CN117321417A - Method for determining the efficacy of therapeutic cell compositions - Google Patents

Method for determining the efficacy of therapeutic cell compositions Download PDF

Info

Publication number
CN117321417A
CN117321417A CN202280036193.9A CN202280036193A CN117321417A CN 117321417 A CN117321417 A CN 117321417A CN 202280036193 A CN202280036193 A CN 202280036193A CN 117321417 A CN117321417 A CN 117321417A
Authority
CN
China
Prior art keywords
recombinant receptor
cells
therapeutic
dependent activity
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280036193.9A
Other languages
Chinese (zh)
Inventor
N·海格
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Juno Therapeutics Inc
Original Assignee
Juno Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Juno Therapeutics Inc filed Critical Juno Therapeutics Inc
Publication of CN117321417A publication Critical patent/CN117321417A/en
Pending legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • G01N33/505Cells of the immune system involving T-cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/566Immunoassay; Biospecific binding assay; Materials therefor using specific carrier or receptor proteins as ligand binding reagents where possible specific carrier or receptor proteins are classified with their target compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70514CD4
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70503Immunoglobulin superfamily, e.g. VCAMs, PECAM, LFA-3
    • G01N2333/70517CD8
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Chemical & Material Sciences (AREA)
  • Urology & Nephrology (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Toxicology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The present disclosure relates to methods of determining the efficacy of therapeutic cell compositions in combination with cell therapies. The cells of the therapeutic cell composition may express recombinant receptors, such as chimeric receptors, e.g., chimeric Antigen Receptors (CARs), or other transgenic receptors, such as T Cell Receptors (TCRs). The methods provide assays for identifying the efficacy of therapeutic cell compositions, including relative efficacy.

Description

Method for determining the efficacy of therapeutic cell compositions
Cross Reference to Related Applications
The present application claims priority from U.S. provisional application 63/164,527 filed on month 22 of 2021, the contents of which are incorporated by reference in their entirety for all purposes.
Incorporated by reference into the sequence listing
The present application is filed with a sequence listing in electronic format. The sequence listing is provided in a file created by month 21 of 2022, titled 73504_2023040_seqlist.txt, which is 84,579 bytes in size. The information of the sequence listing in electronic format is incorporated in its entirety by reference.
Technical Field
The present disclosure relates to methods of determining the efficacy of therapeutic cell compositions for use in conjunction with cell therapies. The cells of the therapeutic cell composition may express recombinant receptors, such as chimeric receptors, e.g., chimeric Antigen Receptors (CARs), or other transgenic receptors, such as T Cell Receptors (TCRs). The methods provide assays for determining the efficacy of therapeutic cell compositions, including relative efficacy.
Background
A variety of immunotherapies and/or cell therapies may be used to treat diseases and conditions. For example, adoptive cell therapies including those involving administration of cells expressing chimeric receptors (e.g., chimeric Antigen Receptors (CARs)) and/or other recombinant antigen receptors specific for a disease or disorder of interest, as well as other adoptive immune cell therapies and adoptive T cell therapies, may be beneficial in the treatment of cancer or other disease or disorder. There is a need for improved methods for characterizing effective therapeutic cell compositions (e.g., in combination with methods for ex vivo production of compositions) and treating subjects with cell therapies. Methods to address such needs are provided herein.
Disclosure of Invention
Provided herein is a method of determining the efficacy of a therapeutic cell composition, the method comprising performing a plurality of incubations, each of the plurality of incubations comprising incubating a cell of a therapeutic cell composition with a recombinant receptor stimulator, the therapeutic composition comprising a cell engineered to express a recombinant receptor, wherein binding of the recombinant receptor stimulator to the recombinant receptor stimulates recombinant receptor-dependent activity of the cell; and each of the plurality of incubations comprises a different stepwise adjustment ratio of cells of the therapeutic cell composition to the recombinant receptor stimulator; measuring recombinant receptor-dependent activity from each of the plurality of incubations; and determining a stepwise adjustment ratio resulting in a specific recombinant receptor-dependent activity, e.g. a half-maximal recombinant receptor-dependent activity, based on the measured recombinant receptor-dependent activity from each of the plurality of incubations. In some of any of the provided embodiments, the method further comprises determining the relative efficacy of the therapeutic cell composition by comparing the stepwise adjustment ratio of the specific receptor-dependent activity (e.g., half-maximal recombinant receptor-dependent activity) resulting in the therapeutic cell composition to the stepwise adjustment ratio of the specific receptor-dependent activity (e.g., half-maximal recombinant receptor-dependent activity) resulting in the reference standard.
Also provided herein is a method of determining the efficacy of a therapeutic cell composition, the method comprising performing a plurality of incubations, each of the plurality of incubations comprising incubating cells of a therapeutic cell composition comprising cells engineered to express a recombinant receptor with a recombinant receptor stimulator, wherein: binding of the recombinant receptor stimulator to the recombinant receptor stimulates recombinant receptor-dependent activity of the cell; and each of the plurality of incubations comprises a different stepwise adjustment ratio of cells of the therapeutic cell composition to the recombinant receptor stimulator; measuring recombinant receptor-dependent activity from each of the plurality of incubations; and determining the relative efficacy of the therapeutic cell composition by comparing the specific recombinant receptor-dependent activity of the therapeutic cell composition (e.g., the half-maximal recombinant receptor-dependent activity of the therapeutic cell composition) to the specific recombinant receptor-dependent activity of a reference standard (e.g., the half-maximal recombinant receptor-dependent activity).
In some any of the provided embodiments, each of the plurality of incubations comprises culturing a constant number of cells of the therapeutic composition with different amounts of the recombinant receptor stimulator to generate a plurality of different stepwise adjustment ratios. In some any of the provided embodiments, each of the plurality of incubations comprises culturing a constant amount of a binding molecule (e.g., a recombinant receptor stimulator) with a different number of cells of the therapeutic composition to generate a plurality of different stepwise adjustment ratios.
In some any of the provided embodiments, the plurality of incubations is performed on two or more, optionally 3, 4, 5, 6, 7, 8, 9, 10 or more therapeutic cell compositions. In some of any of the provided embodiments, the two or more therapeutic cell compositions each comprise the same recombinant receptor. In some of any of the provided embodiments, the two or more therapeutic cell compositions each comprise a different recombinant receptor. In some of any of the provided embodiments, at least one of the two or more therapeutic cell compositions comprises a recombinant receptor that is different from the other therapeutic compositions.
In some of any of the provided embodiments, the two or more therapeutic cell compositions are each manufactured using the same manufacturing process. In some of any of the provided embodiments, the two or more therapeutic cell compositions are each manufactured using a different manufacturing process. In some of any of the provided embodiments, at least one of the two or more therapeutic cell compositions is manufactured using a manufacturing process that is different from the manufacturing process used to manufacture the other therapeutic cell compositions.
In some of any of the provided embodiments, the two or more therapeutic cell compositions are produced by cells from a single subject. In some of any of the provided embodiments, the two or more therapeutic cell compositions are produced by cells from different subjects. In some any of the provided embodiments, the subject is a healthy subject or a subject with a disease or disorder.
In some of any of the provided embodiments, each of the different subjects has the same disease or disorder. In some of any of the provided embodiments, each of the different subjects is to be treated with the same therapeutic cell composition to treat the disease or disorder in the subject.
In some any of the provided embodiments, the plurality of incubations is at least three incubations. In some any of the provided embodiments, the plurality of incubations is at least five incubations. In some any of the provided embodiments, the plurality of incubations is at least seven incubations. In some any of the provided embodiments, the plurality of incubations is at least ten incubations.
In some any of the provided embodiments, the recombinant receptor-dependent activity comprises one or more of: cytokine expression, cytolytic activity, receptor up-regulation, receptor down-regulation, proliferation, gene up-regulation, gene down-regulation, or cellular health. In some of any of the provided embodiments, the recombinant receptor-dependent activity comprises or is cytokine expression or production. In some any of the provided embodiments, the recombinant receptor-dependent activity comprises or is the expression or production of a cytokine, wherein the cytokine is TNF- α, IFN- γ (IFNg), or IL-2. In some of any of the provided embodiments, the recombinant receptor-dependent activity comprises or is a cytolytic activity. In some of any of the provided embodiments, the recombinant receptor-dependent activity comprises or is receptor down-regulation. In some of any of the provided embodiments, the recombinant receptor-dependent activity comprises or is proliferation. In some of any of the provided embodiments, the recombinant receptor-dependent activity comprises or is gene up-regulation. In some of any of the provided embodiments, the recombinant receptor-dependent activity comprises or is down-regulated. In some of any of the provided embodiments, the recombinant receptor-dependent activity comprises or is cellular health. In some any of the provided embodiments, the recombinant receptor-dependent activity comprises or is a cellular health, wherein the cellular health comprises one or more of cell death, cell diameter, viable cell concentration, and cell count.
In some of any of the provided embodiments, the recombinant receptor-dependent activity measured at each of the plurality of incubations is normalized relative to a maximum receptor-dependent activity measured for the therapeutic cell composition.
In some of any of the provided embodiments, the reference standard is a therapeutic cell composition comprising a validated step-by-step adjustment ratio that results in a particular (e.g., half-maximal) recombinant receptor-dependent activity, a commercially available therapeutic cell composition, a therapeutic cell composition manufactured using the same manufacturing process as that used to manufacture the therapeutic cell composition, a therapeutic cell composition manufactured using a manufacturing process different from that used to manufacture the therapeutic cell composition, a therapeutic cell composition comprising the same recombinant receptor as that therapeutic cell composition, a therapeutic cell composition comprising a different recombinant receptor than that therapeutic cell composition, a therapeutic cell composition manufactured from the same subject, or a therapeutic cell composition manufactured from a different subject.
In some any of the provided embodiments, the reference standard is one of the two or more therapeutic compositions.
In some of any of the provided embodiments, the recombinant receptor stimulant comprises a target antigen of the recombinant receptor or an extracellular domain-binding portion thereof, optionally a recombinant antigen. In some of any of the provided embodiments, the recombinant receptor stimulant comprises an ectodomain binding portion of the antigen, and the ectodomain binding portion comprises an epitope recognized by the recombinant receptor. In some of any of the provided embodiments, the recombinant receptor stimulant comprises an antibody that is specific for an extracellular domain (e.g., an epitope on an extracellular domain) of the recombinant receptor. In some of any of the provided embodiments, the recombinant receptor stimulant is an anti-idiotype antibody specific for an extracellular antigen-binding domain of the recombinant receptor.
In some of any of the provided embodiments, the recombinant receptor stimulant is immobilized or attached to a solid support. In some any of the provided embodiments, the solid support is a surface of a vessel, optionally a well of a microplate, in which the plurality of incubations is performed. In some any of the provided embodiments, the solid support is a bead.
In some of any of the provided embodiments, the recombinant receptor stimulant is an antigen-expressing cell, optionally wherein the cell is a clone, a cell from a cell line, or a primary cell taken from a subject. In some any of the provided embodiments, the antigen expressing cell is a cell line. In some any of the provided embodiments, the cell line is a tumor cell line. In some of any of the provided embodiments, the antigen-expressing cell is a cell that has been introduced, optionally by transduction, to express an antigen of the recombinant receptor.
In some of any of the provided embodiments, the stepwise adjustment ratio reaches a linear dose response range of recombinant receptor-dependent activity of the reference standard. In some of any of the provided embodiments, the stepwise adjustment ratio comprises a lower asymptote (minimum) recombinant receptor-dependent activity and a higher asymptote (maximum) recombinant receptor-dependent activity of the reference standard.
In some of any of the provided embodiments, the therapeutic cell composition comprises a single cell subtype enriched or purified from a biological sample or a mixed population of cell subtypes obtained by optionally mixing cell subtypes enriched or purified from a biological sample. In some any of the provided embodiments, the biological sample comprises a whole blood sample, a buffy coat sample, a Peripheral Blood Mononuclear Cell (PBMC) sample, an unfractionated cell sample, a lymphocyte sample, a leukocyte sample, an apheresis product, or a leukocyte apheresis product.
In some any of the provided embodiments, the therapeutic cell composition comprises a primary cell. In some of any of the provided embodiments, the therapeutic cell composition comprises autologous cells from the subject to be treated. In some any of the provided embodiments, the therapeutic cell composition comprises allogeneic cells. In some any of the provided embodiments, the therapeutic cell composition comprises cd3+, cd4+, and/or cd8+ T cells. In some any of the provided embodiments, the therapeutic cell composition comprises or is a cd4+ T cell. In some any of the provided embodiments, the therapeutic cell composition comprises or is a cd8+ T cell. In some any of the provided embodiments, the recombinant receptor is a Chimeric Antigen Receptor (CAR).
In some any of the provided embodiments, the therapeutic cell composition comprises cd4+ T cells and cd8+ T cells. In some any of the provided embodiments, the recombinant receptor is a Chimeric Antigen Receptor (CAR).
In some of any of the provided embodiments, the plurality of incubations is performed in a flask, tube, or multi-well plate. In some of any of the provided embodiments, each of the plurality of incubations is performed separately in a well of a multi-well plate. In some any of the provided embodiments, the multi-well plate is a 96-well plate, a 48-well plate, a 12-well plate, or a 6-well plate.
In some any of the provided embodiments, the method further comprises determining a dose of cells of the therapeutic composition for administration to a subject in need thereof based on the stepwise adjustment ratio resulting in a specific (e.g., half-maximal) recombinant receptor-dependent activity. In some any of the provided embodiments, the method further comprises determining a dose of cells of the therapeutic composition for administration to a subject in need thereof based on the relative potency.
In some any of the provided embodiments, the subject has a disease or disorder. In some any of the provided embodiments, the disease or disorder is cancer.
In some of any of the provided embodiments, the method further comprises determining a manufacturing process that produces an optimal therapeutic cellular composition efficacy based on the relative efficacy, wherein the optimal therapeutic cellular composition efficacy is associated with a complete and/or sustained response and/or reduced toxicity.
In some of any of the provided embodiments, the method further comprises determining a manufacturing process that produces a therapeutic cell composition having a reduced or low difference in potency based on the relative potency, wherein the reduced or low difference is determined by comparison with a difference in a different manufacturing process.
Drawings
FIGS. 1A-1B show cytokine secretion response curves for three different donors at different target to effector cell ratios (T: E). Fig. 1A shows the original cytokine secretion curve and fig. 1B shows the same secretion curve normalized by the higher asymptote (Vmax).
Detailed Description
Provided herein are methods for assessing or determining the efficacy of therapeutic cell compositions (e.g., therapeutic cell compositions) for cell therapies, including engineered T cell therapies, such as for use in conjunction with monitoring ex vivo processes for generating cell therapies and for determining dosages for treating diseases and disorders, including various cancers. The embodiments provided relate to therapeutic T cell compositions containing engineered T cells, such as those engineered to express recombinant proteins, such as those expressing recombinant receptors designed to recognize and/or specifically bind to molecules associated with the disease or disorder and elicit a response, such as an immune response, against such molecules upon binding to such molecules. Receptors can include chimeric receptors, such as Chimeric Antigen Receptors (CARs); and other transgenic antigen receptors, including transgenic T Cell Receptors (TCRs).
The methods of the invention for determining the efficacy of a therapeutic cell composition typically use maximum antigen stimulation of the engineered cells of the therapeutic cell composition. For example, various prior methods measure antigen-specific activity of cells, such as cytokine expression, receptor up-regulation, or down-regulation of therapeutic cell compositions after maximum stimulation. The measured activity in response to the stimulus of all tested therapeutic cell compositions is then compared to determine which therapeutic cell composition has the greatest activity, e.g., recombinant receptor-dependent activity. Therapeutic cell compositions with the highest activity may be considered the most effective therapeutic cell compositions.
In many cases, the use of saturated levels of antigen may not be physiologically relevant. Furthermore, the use of saturated levels of antigen does not capture the sensitivity of therapeutic cell compositions to stimulation by recombinant receptors. For example, current assays do not distinguish between the sensitivity of recombinant receptors to stimuli (e.g., the amount or concentration of antigen) required to elicit a detectable active response. Maximum antigen stimulation also does not allow elucidation of the activity of recombinant receptors against different stimuli, e.g. recombinant receptor-dependent activity. Briefly, current methods of assessing the efficacy of therapeutic cell compositions provide a one-dimensional view of the efficacy of therapeutic cell compositions, and further lack the ability to establish a measure that can capture the sensitivity (e.g., behavior, activity) of the therapeutic cell compositions.
The methods provided herein are designed to more fully assess the sensitivity (e.g., behavior, activity) of therapeutic cell compositions. The methods provided herein are designed to provide a more biologically relevant measure of the efficacy of therapeutic cell compositions. In some embodiments, the efficacy of a therapeutic cell composition determined according to the methods described herein can be more closely related to the safety and efficacy of the therapeutic cell composition. In some embodiments, the efficacy of therapeutic cell compositions determined according to the methods described herein may provide a measure of improved manufacturing control and/or variability, which in turn may allow for improved assessment of stability and activity (e.g., recombinant receptor-dependent activity) of the manufactured therapeutic cell compositions.
The provided methods relate to a direct way to compare how therapeutic cell compositions respond to antigens. Unlike prior methods of comparing the activity against a single target antigen stimulus (in many cases the most likely stimulus), the provided methods stepwise adjust the ratio of targets (e.g., antigen or antibody expressing cells) to effector cells (cells of a therapeutic composition). For example, this ratio can be controlled by maintaining a constant number of effector cells in the assay and by varying the number of target expressing cells. For example, by the provided methods, the number of target-expressing cells and/or the amount of target (e.g., antigen or antibody) required to achieve a particular receptor-dependent activity can be determined. In some embodiments, the target is an antigen of a recombinant receptor. Thus, in some cases, the target expressing cell is an antigen expressing cell. In some embodiments, the specific receptor-dependent activity is 50% of the maximum activity. In some embodiments, the specific receptor-dependent activity is 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80% or 90% of the maximum receptor-dependent activity. In some embodiments, the specific receptor-dependent activity is within the scope as disclosed herein. In some embodiments, the provided methods allow for comparison of therapeutic cell compositions. For example, therapeutic cell compositions can be assessed by the methods provided herein and relative efficacy determined, for example, as described herein.
It is important to know whether it is possible to compare the activity between different therapeutic cell compositions. In particular, the ability to compare therapeutic cell compositions is unknown because it is believed that therapeutic cell compositions have different sensitivity characteristics (e.g., response to recombinant receptor-specific stimuli), such as different minimum and maximum responses. Variables affecting therapeutic cell compositions are much larger than other pharmaceutical products (e.g., biological agents), such as due to differences in: the donor from which the cells are derived or obtained, the cell (e.g., differentiated state), the antigen binding capacity of a particular recombinant receptor (e.g., CAR), the intracellular signaling component of a particular recombinant receptor, the percentage or frequency of cells in the composition expressing the recombinant receptor (e.g., CAR), the process used to produce the therapeutic composition, and other factors. For example, as shown in fig. 1A, therapeutic compositions from different donors exhibited different responses to antigen stimulation, as shown by the response curves. Thus, this result would indicate that it may not be possible to compare therapeutic cell compositions. It was found herein that by normalizing the higher asymptote (e.g., maximum stimulus) of the activity relative to the response curve, the sensitivity of therapeutic cell compositions can be compared. The provided methods enable the measurement of sensitivity between different cell products, including those that may vary, such as due to production by different methods, expression of different antigen receptors, production from different donors, and other variables. The existing method evaluates activity at maximum stimulation, in part because it is considered the only method to account for potential differences.
In some embodiments, the methods provided herein reduce or eliminate sources of variability. For example, the methods provided herein are robust to variability that may occur due to donor heterogeneity and/or daily sampling or testing. In some cases, eliminating variability (e.g., variability due to donor heterogeneity and/or sampling or testing) allows for comparison of therapeutic cell compositions.
The methods provided herein include assay formats comprising a series of incubations in which cells of therapeutic cell compositions are incubated with recombinant receptor stimulators at different stepwise adjustment ratios. In a provided aspect, the recombinant receptor stimulant is an agent that induces or is capable of inducing a signal through an intracellular signaling region of a recombinant receptor. For example, a recombinant receptor stimulant may include an antigen of a recombinant receptor, such as a purified antigen or recombinant antigen, an antigen-expressing cell, or an anti-idiotype antibody that has specificity for an extracellular antigen-binding domain (e.g., scFv) of the recombinant receptor. In some embodiments, the methods provided herein (including assay formats) are designed to measure the sensitivity of a therapeutic cell composition by measuring or determining the amount or concentration of a recombinant receptor stimulator (e.g., as described in section I-B) required to stimulate recombinant receptor-dependent activity of an engineered cell of the therapeutic cell composition. For example, the methods provided herein can determine the level (e.g., amount, concentration) of an antigen for stimulating a quantifiable and detectable activity (e.g., recombinant receptor-dependent activity) of a therapeutic cell composition. In some embodiments, the measurement of sensitivity includes measurement of recombinant receptor-dependent activity stimulated by binding of the recombinant receptor stimulator to the recombinant receptor across multiple stepwise adjustment ratios. The ability of the method to assess recombinant receptor-dependent activity at different stepwise adjustment ratios allows for the determination, estimation and/or extrapolation of the general activity or behavior of therapeutic cell compositions against recombinant receptor-specific stimuli.
The methods provided herein include assays that allow for assessing the efficacy of a therapeutic cell composition by measuring the activity of cells of the therapeutic cell composition expressing a recombinant receptor (e.g., a recombinant receptor described herein) in response to stimulation of the recombinant receptor over a series of controlled incubations. For example, a series of incubations can include incubating engineered cells of a therapeutic cell composition expressing a recombinant receptor with a recombinant receptor stimulator (e.g., as described herein (e.g., section I-B)), which when bound to the recombinant receptor, stimulates activity of the recombinant receptor expressed by the cell, e.g., recombinant receptor-dependent activity, at different stepwise adjustment rates, wherein each incubation is a different stepwise adjustment rate. In some embodiments, incubating the cells or at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more, each incubating the cells containing a different ratio of therapeutic cell composition with the recombinant receptor stimulant is performed. In some embodiments, the incubating is performed with cells containing a different stepwise adjustment ratio of the therapeutic cell composition to the recombinant receptor stimulant, or with at least 3 incubations. In some embodiments, a single or at least 6 incubations are performed, each incubation containing cells of a different stepwise adjustment ratio of therapeutic cell composition to recombinant receptor stimulant. In some embodiments, the incubating is performed with cells containing a different stepwise adjustment ratio of the therapeutic cell composition to the recombinant receptor stimulant, or with at least 10 incubations.
In some embodiments, a constant number of cells of the therapeutic composition are incubated with different amounts of the recombinant receptor stimulant to generate a series (e.g., a plurality) of different stepwise adjustment ratios. Alternatively, in some embodiments, a constant amount or concentration of recombinant receptor stimulant (e.g., as described in section I-B) may be incubated with different numbers of cells of the therapeutic cell composition to generate a series (e.g., a plurality) of different stepwise adjustment ratios. Regardless of how different stepwise adjustment ratios are achieved (e.g., by varying the total number of cells of the therapeutic cell composition or the amount of recombinant receptor stimulating agent), the use of a series (e.g., a plurality) of stepwise adjustment ratios allows for the assessment of recombinant receptor-dependent activity across a range of stimulation conditions. In some embodiments, the measurement range can be used to extract, estimate, and/or determine how engineered cells of a particular therapeutic cell composition respond to different levels of recombinant receptor stimulation.
Any number of metrics may be determined, extracted, extrapolated, estimated, and/or inferred from the measured recombinant receptor-dependent activity produced according to the methods described herein. Non-limiting examples of metrics include a gradual adjustment ratio at which maximum, minimum, and half maximum (50%) recombinant receptor-dependent activity occurs, a gradual adjustment ratio at which a particular percentage (e.g., 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, or 90%) of maximum recombinant receptor-dependent activity occurs, and a gradual adjustment ratio that encompasses a range of recombinant receptor-dependent activities (e.g., 10% -90%, 20% -80%, 30% -70%, 40% -60% of maximum recombinant receptor-dependent activity). In some embodiments, the measured recombinant receptor-dependent activity of the therapeutic cell composition is curve fitted to generate a recombinant receptor-dependent activity curve. In some embodiments, the curve is similar to a dose response curve. In some embodiments, a measure of recombinant receptor-dependent activity and/or a ratio at which a particular recombinant receptor-dependent activity occurs is extrapolated and/or estimated from the curve. In some embodiments, the recombinant receptor-dependent activity curve may be used to extrapolate values or measures for therapeutic cell compositions and/or recombinant receptor stimulators at which a particular recombinant receptor-dependent activity occurs. In some embodiments, for example, when the therapeutic cell composition cell count remains constant and the amount of recombinant receptor stimulant varies, the amount (e.g., mass (in picograms) in the case of a purified or recombinant target or cell number in the case of a target expressing cell) or concentration (e.g., mass/volume (e.g., in pg/mL) in the case of a purified or recombinant target or cell number per unit volume (e.g., individual cells/mL) in the case of a target expressing cell) of the recombinant receptor stimulant is used to determine the maximum, minimum, half-maximum, and range at which recombinant receptor-dependent activity occurs. In some embodiments, for example, when the cell count of the therapeutic cell composition changes and the amount of recombinant receptor stimulant remains constant, the cell number (e.g., count, total number) of the therapeutic cell composition is used to determine the maximum, minimum, half-maximum, and range at which recombinant receptor-dependent activity occurs. In some embodiments, one or more of the following is used to determine a stepwise adjustment of the ratio maxima, minima, half maxima and ranges at which the recombinant receptor-dependent activity occurs. In some embodiments, the amount or concentration of recombinant receptor stimulant is used to determine the half maximal recombinant receptor-dependent activity. In some embodiments, the amount (e.g., count) of cells of the therapeutic cell composition is used to determine the half maximal recombinant receptor-dependent activity. In some embodiments, stepwise adjustment of the ratio is used to determine half maximal recombinant receptor-dependent activity. These exemplary metrics, as well as other metrics not listed, provide a quantitative description of the therapeutic cell composition, which can be used to determine the efficacy and/or relative efficacy of the therapeutic cell composition (e.g., efficacy relative to a reference standard as described herein (e.g., section I-D)).
In some embodiments, the efficacy of a therapeutic cell composition is expressed as a value or measure of a stepwise adjustment ratio determined based on recombinant receptor-dependent activity, the amount of cells of the therapeutic cell composition, and/or the amount or concentration of recombinant receptor stimulant. In some embodiments, the efficacy of the therapeutic cell composition is a value or measure of the stepwise adjustment ratio at which half-maximum (e.g., 50% of maximum activity) of the recombinant receptor-dependent activity occurs, the amount of cells of the therapeutic cell composition, and/or the amount or concentration of the recombinant receptor stimulator. In some embodiments, the potency of the therapeutic cell composition is a stepwise adjustment ratio at which half maximum of the recombinant receptor-dependent activity occurs (e.g., 50% of maximum activity). In some embodiments, the potency of the therapeutic cell composition is the concentration of the recombinant receptor stimulant at which half-maximum of the recombinant receptor-dependent activity occurs. In some embodiments, the half-maximum of the recombinant receptor-dependent activity reflects a step-wise adjustment ratio at which 50% effective stimulation (ES 50) of the therapeutic cell composition occurs, the concentration of the recombinant receptor stimulator, and/or the cell count, as measured by the recombinant receptor-dependent activity.
In some embodiments, the potency of the therapeutic cell composition is relative potency. For example, the stepwise adjustment ratio at which the half maximal recombinant receptor-dependent activity of the therapeutic cell composition is measured can be compared to the stepwise adjustment ratio at which the half maximal recombinant receptor-dependent activity of the reference standard is measured. It will be appreciated that the concentration or amount of recombinant receptor stimulant or cell count may be used instead of stepwise adjustment of the ratio, if applicable. In some embodiments, the reference standard is a therapeutic cell composition having a known and/or validated step-by-step adjustment ratio at which half-maximal recombinant receptor-dependent activity occurs. In some embodiments, the reference standard is a commercially available therapeutic cell composition, for example, whose stepwise adjustment ratio (at which half maximum recombinant receptor-dependent activity occurs) has been determined using the methods as described herein. In some embodiments, the reference standard is a different therapeutic cell composition, for example, whose stepwise adjustment ratio (at which half maximum recombinant receptor-dependent activity occurs) has been determined using the methods as described herein. In some embodiments, the different therapeutic cell compositions contain cells that express recombinant receptors that bind the same antigen as the test therapeutic cell composition but have different receptor structures. In some embodiments, the different therapeutic cell composition contains cells that express the same recombinant receptor as the test therapeutic cell composition, but the therapeutic cell composition is manufactured using a different process than that used to manufacture the test therapeutic cell composition. In some embodiments, the relative efficacy is a ratio determined by dividing the stepwise adjustment ratio resulting in a half maximum of the test therapeutic cell composition by the stepwise adjustment ratio resulting in a half maximum of the reference standard. In some embodiments, the relative efficacy is a percentage determined by dividing the step-wise adjustment ratio that results in a half-maximum of the test therapeutic cell composition by the step-wise adjustment ratio that results in a half-maximum of the reference standard and multiplying by 100.
In some cases, normalizing the recombinant receptor-dependent activity of a therapeutic cell composition can be used to determine whether the recombinant receptor-dependent activity of two or more therapeutic cell compositions can be compared. For example, if recombinant receptor-dependent activity of two or more therapeutic compositions is determined and the maximum and/or minimum recombinant receptor-dependent activity of each therapeutic cell composition tested is different, normalizing the recombinant receptor-dependent activity of each composition relative to its own maximum value may allow for an assessment of the suitability of comparing the recombinant receptor-dependent activities.
In some embodiments, the recombinant receptor-dependent activity is normalized to a measured maximum recombinant receptor-dependent activity value. In some embodiments, the recombinant receptor-dependent activity curve is normalized to the measured maximum recombinant receptor-dependent activity. In some embodiments, when the recombinant receptor-dependent activity curve is normalized, the maximum activity value is the average of the higher asymptotes of the curve. In some embodiments, normalizing the recombinant receptor-dependent activity of the therapeutic cell composition and the reference standard relative to their respective maxima facilitates a comparison between the test therapeutic cell composition and the reference standard. In some embodiments, normalizing the recombinant receptor-dependent activity of the therapeutic cell composition and the reference standard relative to their respective maxima facilitates calculation of relative potency.
In some embodiments, normalizing the recombinant receptor-dependent activity of the therapeutic cell composition and the reference standard relative to their respective maxima allows for parallel line testing. In some embodiments, the results of the parallel line test are indicative of the ability to compare the therapeutic cell composition to a reference standard.
The methods provided herein for assessing the efficacy of therapeutic cell compositions (including assays) allow for comparison of different therapeutic cell compositions (including reference standards). The ability to compare therapeutic cell compositions provides a method for identifying not only therapeutic cell compositions with improved, optimal and/or consistent efficacy, but also for: identifying candidate therapeutic cell compositions for further development and/or analysis; identifying manufacturing processes and procedures that produce therapeutic cell compositions with improved or optimal efficacy; identifying a manufacturing procedure or process that produces a therapeutic cell composition with consistent efficacy and/or estimates variability inherent to the manufacturing procedure; determining a dose of a therapeutic cell composition to be administered to a subject in need thereof, e.g., a dose that will produce a clinical response without producing toxicity; and/or comparing the efficacy of the allogeneic therapeutic cell composition with the autologous therapeutic cell composition. The methods provided herein are designed to be compatible with a relative potency format that is agnostic to whether the test therapeutic composition or reference standard is from a different donor (e.g., subject), manufacturing process, and/or therapeutic product.
All publications (including patent documents, scientific articles, and databases) mentioned in this application are incorporated by reference in their entirety for all purposes to the same extent as if each individual publication was individually incorporated by reference. If the definition set forth herein is contrary to or otherwise inconsistent with the definition set forth in the patents, applications, published applications and other publications incorporated by reference, the definition set forth herein takes precedence over the definition incorporated by reference.
The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.
I. Cellular potency assay
Provided herein are methods of assessing the efficacy of a therapeutic cell composition, e.g., a therapeutic T cell composition (e.g., as described in section I-a) comprising T cells (e.g., cd3+, cd4+, cd8+ T cells) engineered to express a recombinant receptor (e.g., CAR), using an assay comprising a plurality of incubations, wherein each of the plurality of incubations comprises incubating cells of the therapeutic cell composition comprising cells engineered to express a recombinant receptor with a recombinant receptor stimulator (e.g., an antigen, antigen-expressing cell, or antibody (e.g., an anti-idiotype antibody)) as described, e.g., in section I-B, that is recognized by the recombinant receptor or is capable of being bound by the recombinant receptor to stimulate recombinant receptor-dependent activity of the cell. In some embodiments, the recombinant receptor-dependent activity is an activity of a cell elicited in response to stimulation of its recombinant receptor. In some embodiments, recombinant receptor-dependent activity is an activity such as, for example, cytokine expression, cytolytic activity, receptor up-or down-regulation, gene up-or down-regulation, cytolytic activity, proliferative activity, and/or a measure of cell health, e.g., as described in section I-C. In some embodiments, each of the plurality of incubations contains a different step-wise adjustment ratio of cells of the therapeutic cell composition to recombinant receptor stimulant. In some embodiments, each of the plurality of incubations comprises incubating a constant number of cells of the therapeutic composition with a different amount of the recombinant receptor stimulator to generate a plurality of different stepwise adjustment ratios. In some embodiments, each of the plurality of incubations comprises incubating a different number of cells of the therapeutic composition with a constant amount or concentration of the recombinant receptor stimulator to generate a plurality of different stepwise adjustment ratios. In some embodiments, incubating the cells or at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more, each incubating the cells containing a different ratio of therapeutic cell composition with the recombinant receptor stimulant is performed. In some embodiments, the incubating is performed in a single or at least 3 incubations, each incubation containing a different ratio of cells of the therapeutic cell composition to recombinant receptor stimulant. In some embodiments, the incubation is performed for at least 6 incubations, each incubation containing a different ratio of cells of the therapeutic cell composition to recombinant receptor stimulant. In some embodiments, the incubation is performed for at least 10 or more incubations, each incubation containing a different ratio of cells of the therapeutic cell composition to recombinant receptor stimulant. In some embodiments, recombinant receptor-dependent activity from each of the plurality of incubations is measured. In some embodiments, recombinant receptor-dependent activity from each of the plurality of incubations is measured, e.g., by fluorescence, flow cytometry, ELISA, as described in section I-C. In some embodiments, the measurement of the recombinant receptor-dependent activity is fitted by a curve to produce a recombinant receptor-dependent activity curve, e.g., as described above. In some embodiments, the stepwise adjustment ratio resulting in half-maximal recombinant receptor-dependent activity is determined based on recombinant receptor-dependent activity measured from each of the plurality of incubations. In some embodiments, the stepwise adjustment ratio resulting in half-maximal recombinant receptor-dependent activity is extrapolated, or estimated from a recombinant receptor-dependent activity curve. In some embodiments, the recombinant receptor-dependent activity curve is normalized to the measured maximum recombinant receptor-dependent activity. In some embodiments, the gradual adjustment ratio that results in half-maximal recombinant receptor-dependent activity is the efficacy of the therapeutic cell composition. In some embodiments, instead of stepwise adjusting the ratio, the concentration or amount of recombinant receptor stimulant or the cell count may be reported, if applicable.
In some embodiments, the stepwise adjustment ratio resulting in half maximal recombinant receptor-dependent activity is compared to the stepwise adjustment ratio resulting in half maximal recombinant receptor-dependent activity of a reference standard. For example, the stepwise adjustment ratio of a therapeutic cell composition that results in half-maximal recombinant receptor-dependent activity is divided by the stepwise adjustment ratio of half-maximal recombinant receptor-dependent activity that results in a reference standard (e.g., determined according to the methods described herein) to produce relative potency. In some embodiments, the relative efficacy is expressed as a ratio. In some embodiments, the relative efficacy is expressed as a percentage.
The methods provided herein for determining efficacy may be performed repeatedly. For example, the assay may be performed 2, 3, 4, 5 or more times. In some embodiments, repeated experiments are used to confirm the accuracy and/or precision of the assay, including the measured recombinant receptor-dependent activity and/or the consistency of the determined potency and/or relative potency. In some embodiments, a single assay is performed by assaying for a particular therapeutic cell composition in duplicate or triplicate. In some embodiments, the assay is performed in duplicate. In some embodiments, the assay is performed in triplicate. In some cases, such as where the assay is performed in duplicate or triplicates, the measured recombinant receptor-dependent activity from each replicate experiment is used to provide a statistical measure of the recombinant receptor-dependent activity. For example, in some cases, the mean, median, standard deviation, and/or variance of each measure of recombinant receptor-dependent activity is determined. In some embodiments, the average value of each measure of recombinant receptor-dependent activity is determined. In some embodiments, the standard deviation of each measure of recombinant receptor-dependent activity is determined. In some embodiments, the average measure of recombinant receptor-dependent activity is fitted using a mathematical model to generate a recombinant receptor-dependent activity curve. In some embodiments, the curve is normalized to the average maximum. In some embodiments, the average stepwise adjustment ratio that results in half-maximal recombinant receptor-dependent activity is the efficacy of the therapeutic cell composition. In some embodiments, instead of stepwise adjusting the ratio, the average concentration or amount of recombinant receptor stimulant or the cell count may be reported, if applicable. In some embodiments, the potency of the therapeutic cell composition is a relative potency determined by taking the average stepwise adjustment ratio that results in half-maximal recombinant receptor-dependent activity and comparing the average stepwise adjustment ratio to a single or average stepwise adjustment ratio that results in half-maximal recombinant receptor-dependent activity of a reference standard. In some embodiments, the relative potency is the average potency of the therapeutic cell composition divided by the single or average potency of the reference standard. In some embodiments, the relative efficacy is expressed as a ratio. In some embodiments, the relative efficacy is expressed as a percentage.
The assays provided herein can be performed in any one or more vessels suitable for multiple incubations. In some embodiments, the assay is performed in a flask. In some embodiments, the assay is performed in a tube (e.g., microcentrifuge tube, PCR tube, tube). In some embodiments, the assay is performed in a multi-well plate. For example, the multi-well plate is a 6-well plate, a 12-well plate, a 24-well plate, a 48-well plate, or a 96-well plate. In certain embodiments, the assay is performed or carried out in a 12-well plate.
The conditions under which the therapeutic cell composition is incubated with the recombinant receptor stimulant may include one or more of the following: specific media, temperature, oxygen content, carbon dioxide content, time, and/or agents (e.g., nutrients, amino acids, antibiotics, ions). It is contemplated that the duration of the plurality of incubations is at least equivalent to the minimum amount of time for the potential recombinant receptor-dependent activity to be detected (e.g., measured). For example, the amount of time required to accurately measure cytokine expression may be longer than the amount of time required to measure gene expression. It is further contemplated that within one type of activity (e.g., cytokine expression), there may be a temporal difference in the particular cytokine to be measured compared to another. In some embodiments, the plurality of incubations is performed for about, or at least 1, 2, or 3 days. In some embodiments, the plurality of incubations is performed for about, or at least 1 or 2 days. In some embodiments, the plurality of incubations is performed for about, or at least 24, 36, 48, 60, or 72 hours. In some embodiments, the plurality of incubations is performed for about, or at least 24 or 48 hours. In some embodiments, the plurality of incubations is performed for or between about 24 hours and about 72 hours. In some embodiments, the plurality of incubations is performed for or between about 24 hours and about 48 hours. In some embodiments, the plurality of incubations is performed for about, or at least 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, or 60 minutes. In some embodiments, the plurality of incubations is performed for about, or at least 30 minutes. In some embodiments, the plurality of incubations is performed for about, or at least 60 minutes. In some embodiments, the plurality of incubations is performed at or between about 10 and 60 minutes, 20 and 60 minutes, 30 and 60 minutes, 40 and 60 minutes, 50 and 60 minutes.
In some embodiments, the plurality of incubations is performed at a temperature of from about 25 ℃ to about 38 ℃, such as from about 30 ℃ to about 37 ℃, for example, or about 37 ℃ ± 2 ℃. In some embodiments, the plurality of incubations is with from about 2.5% to about 7.5%, such as from about 4% to about 6%, e.g., or about 5% ± 0.5% CO 2 Horizontally. In some embodiments, the plurality of incubations are at a temperature of or about 37 ℃ and/or at a CO of or about 5% 2 Horizontally.
A. Therapeutic cell compositions
The methods provided herein relate to assessing the efficacy of a therapeutic cell composition (e.g., a therapeutic T cell composition) made, for example, by any process. In some embodiments, the methods provided herein can be used to evaluate therapeutic cell compositions made according to the processes described herein (e.g., section II). In some embodiments, the efficacy and/or relative efficacy of a plurality of therapeutic cell compositions made by any of the processes can be assessed according to the methods provided herein. In some embodiments, the plurality of therapeutic cell compositions evaluated are produced by the same manufacturing process. In some embodiments, the plurality of therapeutic cell compositions are manufactured by the same manufacturing process, but comprise different recombinant receptors. In some embodiments, the target is an antigen of a recombinant receptor. Thus, in some cases, the target expressing cell is an antigen expressing cell. In some embodiments, the different recombinant receptors all bind the same target, e.g., target antigen. In some embodiments, different recombinant receptors bind different targets, e.g., target antigens. In some embodiments, the plurality of therapeutic cell compositions evaluated are produced by different manufacturing processes. In some embodiments, the plurality of therapeutic cell compositions are manufactured by different manufacturing processes, but comprise the same recombinant receptor. In some embodiments, the plurality of therapeutic cell compositions are manufactured by different manufacturing processes and comprise different recombinant receptors. In some embodiments, the different recombinant receptors all bind the same antigen. In some embodiments, the plurality of therapeutic cell compositions are manufactured from a single subject. In some embodiments, the plurality of therapeutic cell compositions are manufactured from different subjects. In some embodiments, the subject is a healthy donor. In some embodiments, the subject has a disease or disorder, such as cancer. The methods provided herein can allow for comparison of potency and/or relative potency between therapeutic cell compositions (including as a reference standard for therapeutic cell compositions), regardless of the method of manufacture.
In some embodiments, the therapeutic cell composition is produced or manufactured in conjunction with a process (see, e.g., section II-a) that produces or generates a therapeutic cell composition containing engineered T cells from one or more input populations (e.g., input populations obtained, selected, or enriched from a single biological sample). In certain embodiments, the therapeutic cell composition contains cells that express a recombinant receptor (e.g., CAR, TCR). In certain embodiments, the cells of the therapeutic cell composition are suitable for administration to a subject as a therapy (e.g., autologous cell therapy, allogeneic cell therapy). The methods provided herein can be used to evaluate the efficacy and/or relative efficacy of therapeutic cell compositions for cell therapies.
In some embodiments, the process for generating or producing a therapeutic cell composition of engineered T cells comprises some or all of the following steps: collecting or obtaining a biological sample; isolating, selecting or enriching input cells from a biological sample; freezing and storing at low temperature and then thawing the input cells; selecting and stimulating an input cell of interest, e.g., a T cell, e.g., cd3+, cd4+, cd8+ T cell; genetically engineering the stimulated cells to express or contain a recombinant polynucleotide, e.g., a polynucleotide encoding a recombinant receptor (e.g., CAR); formulating the cultured cells in an output composition; and cryogenically freezing and storing the formulated output cells until the cells are released for infusion and or administration to a subject. In some embodiments, the method of making a therapeutic cell composition does not include a step of expanding cells or increasing the number of cells during the process, such as by incubating the cells in a bioreactor under conditions where the cells expand, e.g., to the following threshold amounts: the amount, level or concentration of cells is at least 2-fold, 3-fold, 4-fold, 5-fold or more compared to the input population. In some embodiments, the method of making a therapeutic cell composition comprises the step of expanding cells or increasing the number of cells during the process, such as by incubating or incubating the cells in a bioreactor under conditions where the cells expand, e.g., to the following threshold amounts: the amount, level or concentration of cells is at least 2-fold, 3-fold, 4-fold, 5-fold or more compared to the input population. In some embodiments, genetically engineering the cells is or includes the step of transducing the cells with a viral vector, such as by: the cells are spin seeded in the presence of the viral particles and then incubated under static conditions in the presence of the viral particles. See, for example, sections II-C.
In certain embodiments, the total duration of the process for generating engineered cells from the initial stimulation to collecting, harvesting, or formulating the cells is about or less than 36 hours, 42 hours, 48 hours, 54 hours, 60 hours, 72 hours, 84 hours, 96 hours, 108 hours, or 120 hours. In some embodiments, the total duration of the provided process for generating engineered cells from the beginning of stimulation to the collection, harvesting, or formulation of cells is between or between about 36 hours and 120 hours, 48 hours and 96 hours, or 48 hours and 72 hours, inclusive. In particular embodiments, the amount of time to complete the provided process is, about or less than 48 hours, 72 hours or 96 hours, as measured from the beginning of incubation to harvesting, collecting or formulating the cells. In particular embodiments, the amount of time to complete the provided process as measured from initial incubation to harvesting, collecting, or formulating the cells is 48 hours±6 hours, 72 hours±6 hours, or 96 hours±6 hours.
In some embodiments, the entire manufacturing process is performed with a single population of enriched T cells (e.g., cd3+, cd4+ and cd8+ T cells). In certain embodiments, the manufacturing process is performed with two or more input populations of enriched T cells that are combined prior to and/or during the process to generate or produce a single therapeutic cell composition of enriched T cells (e.g., a therapeutic cell composition containing cd4+ and cd8+ T cells). In some embodiments, the enriched T cells are or comprise engineered T cells, e.g., T cells transduced to express a recombinant receptor.
In some embodiments, the duration or amount of time required to complete the provided process is, is about or less than 48 hours, 72 hours, 96 hours, 120 hours, 4 days, 5 days, 7 days or 10 days, as measured from the time that the input cells (e.g., cd4+ or cd8+ T cells) are isolated, enriched, and/or selected from the biological sample to the time that the engineered cells of the therapeutic cell composition are collected, formulated, and/or cryoprotected. In some embodiments, the duration or amount of time required to complete the provided process, as measured from the time of isolation, enrichment, and/or selection of the input cells (e.g., cd4+ or cd8+ T cells) from the biological sample to the time of collection, formulation, and/or cryoprotection of the engineered cells, is about 4 to 5 days. In some embodiments, the duration or amount of time required to complete the provided process is or is about 5 days, as measured from the time the input cells (e.g., cd4+ or cd8+ T cells) are isolated, enriched, and/or selected from the biological sample to the time the engineered cells are collected, formulated, and/or cryoprotected. In some embodiments, the duration or amount of time required to complete the provided process is, as measured from the time the input cells (e.g., cd4+ or cd8+ T cells) are isolated, enriched, and/or selected from the biological sample to the time the engineered cells are collected, formulated, and/or cryoprotected, is, less than 5 days. In some embodiments, the duration or amount of time required to complete the provided process is or is about 4 days, as measured from the time the input cells (e.g., cd4+ or cd8+ T cells) are isolated, enriched, and/or selected from the biological sample to the time the engineered cells are collected, formulated, and/or cryoprotected. In some embodiments, the isolated, selected, or enriched cells are not cryogenically protected prior to stimulation, and the duration or amount of time required to complete the provided process is, or is less than, about 48 hours, 72 hours, 96 hours, or 120 hours as measured from the time the cells are isolated, enriched, and/or selected for input to the collection, formulation, and/or cryogenically protected engineered cells.
In certain embodiments, the therapeutic cell composition is made from a population of cells (e.g., cd4+ and cd8+ T cells or cd3+ T cells) isolated, enriched, or selected from a biological sample. In some aspects, the time from when the biological sample is collected from the subject to when the therapeutic cell composition is produced or generated is within a reduced amount of time as compared to other methods or processes.
In some embodiments, at least 30%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, or at least 90%, at least 95% of the cells in the therapeutic cell composition express the recombinant receptor. In certain embodiments, at least 50% of the cells in the therapeutic cell composition express the recombinant receptor. In certain embodiments, at least 30%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95% of the cd3+ T cells in the therapeutic cell composition express the recombinant receptor. In some embodiments, at least 50% of the cd3+ T cells in the therapeutic cell composition express the recombinant receptor. In particular embodiments, at least 30%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 99%, or more than 99% of the cd4+ T cells in the therapeutic cell composition express the recombinant receptor. In certain embodiments, at least 50% of the cd4+ T cells in the therapeutic cell composition express the recombinant receptor. In some embodiments, at least 30%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 97%, at least 99%, or more than 99% of the cd8+ T cells in the therapeutic cell composition express the recombinant receptor. In certain embodiments, at least 50% of the cd8+ T cells in the therapeutic cell composition express the recombinant receptor.
In particular embodiments, the majority of cells of the therapeutic cell composition are naive, central memory, and/or effector memory cells. In certain embodiments, the majority of cells of the therapeutic cell composition are naive or central memory cells. In some embodiments, the majority of cells of the therapeutic cell composition are positive for one or more of CCR7 or CD27 expression. In certain embodiments, the cells of the therapeutic cell composition have a greater share of naive or central memory cells than the output population generated from an alternative process (e.g., a process involving expansion).
In certain embodiments, cells of the therapeutic cell composition have a low fraction and/or frequency of depleted and/or senescent cells. In certain embodiments, the cells of the output population have a low share and/or frequency of depleted and/or senescent cells. In some embodiments, less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, or less than 1% of the cells in the therapeutic cell composition are depleted and/or senescent. In certain embodiments, less than 25% of the cells in the therapeutic cell composition are depleted and/or senescent. In certain embodiments, less than 10% of the cells in the output population are depleted and/or senescent. In certain embodiments, the cells have a low fraction.
In some embodiments, the cells of the therapeutic cell composition have a low share and/or frequency of cells negative for CD27 and CCR7 expression (e.g., surface expression). In particular embodiments, cells of the therapeutic cell composition have a low fraction and/or frequency of CD27-CCR 7-cells. In some embodiments, less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, or less than 1% of the cells in the therapeutic cell composition are CD27-CCR 7-cells. In certain embodiments, less than 25% of the cells in the therapeutic cell composition are CD27-CCR 7-cells. In certain embodiments, less than 10% of the cells in the therapeutic cell composition are CD27-CCR 7-cells. In embodiments, less than 5% of the cells in the therapeutic cell composition are CD27-CCR 7-cells.
In some embodiments, the cells of the therapeutic cell composition have a high share and/or frequency of cells positive for one or both of CD27 and CCR7 expression (e.g., surface expression). In some embodiments, the cells of the therapeutic cell composition have a high share and/or frequency of cells positive for one or both of CD27 and CCR 7. In some embodiments, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or greater than 95% of the cells in the therapeutic cell composition are positive for one or both of CD27 and CCR 7. In various embodiments, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or greater than 95% of the cd4+ car+ cells in the therapeutic cell composition are positive for one or both of CD27 and CCR 7. In some embodiments, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or greater than 95% of the cd8+ car+ cells in the therapeutic cell composition are positive for one or both of CD27 and CCR 7.
In certain embodiments, the cells of the therapeutic cell composition have a high share and/or frequency of cells positive for CD27 and CCR7 expression (e.g., surface expression). In some embodiments, the cells of the therapeutic cell composition have a high fraction and/or frequency of cd27+ccr7+ cells. In some embodiments, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or greater than 95% of the cells in the therapeutic cell composition are cd27+ccr7+ cells. In various embodiments, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or greater than 95% of the cd4+ car+ cells in the therapeutic cell composition are cd27+ ccr7+ cells. In some embodiments, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or greater than 95% of the cd8+ car+ cells in the therapeutic cell composition are cd27+ ccr7+ cells.
In certain embodiments, the cells of the therapeutic cell composition have a low share and/or frequency of cells that are negative for CCR7 expression (e.g., surface expression) and positive for CD45RA expression (e.g., surface expression). In some embodiments, the cells of the therapeutic cell composition have a low fraction and/or frequency of CCR7-cd45ra+ cells. In particular embodiments, less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, or less than 1% of the cells in the therapeutic cell composition are CCR7-cd45ra+ cells. In some embodiments, less than 25% of the cells in the output population (e.g., therapeutic cell composition) are CCR7-cd45ra+ cells. In certain embodiments, less than 10% of the cells in the output population (e.g., therapeutic cell composition) are CCR7-cd45ra+ cells. In certain embodiments, less than 5% of the cells in the therapeutic cell composition are CCR7-cd45ra+ cells.
In some embodiments, the therapeutic cell manufacturing process is different, such that alternative manufacturing processes may be compared, for example, by comparing the efficacy of differently manufactured therapeutic cell compositions. For example, in some embodiments, an alternative process may contain steps for expanding cells. In some embodiments, the alternative process may not contain a step for expanding the cells. In some embodiments, the alternative process includes separate steps for cell selection and stimulation. In some embodiments, the alternative process comprises a single step for cell selection and stimulation. In some embodiments, alternative processes may differ in one or more particular aspects, but otherwise contain similar or identical features, aspects, steps, stages, reagents, and/or conditions of the processes related to the provided methods. In some embodiments, the alternative processes differ in means including, but not limited to, one or more of the following: including different reagent and/or media formulations; the presence of serum during incubation, transduction, transfection and/or incubation; different cell compositions of the input population, e.g., ratio of cd4+ to cd8+ T cells; different stimulation conditions and/or different stimulating agents; different ratios of stimulating agent to cells; different transduction vectors and/or methods; different timing or sequence of incubation, transduction and/or transfection of cells; absence or difference (e.g., different cytokines or different concentrations) of one or more recombinant cytokines present during incubation or transduction; or different timing of harvesting or harvesting of cells.
In some embodiments, the cells of the therapeutic cell composition are engineered to express a recombinant receptor, such as a CAR or TCR (see, e.g., section III), that specifically binds to a ligand, such as a ligand associated with a disease or disorder (e.g., associated with or expressed on a cell of a tumor or cancer). In some embodiments, the recombinant receptor contains an extracellular ligand binding domain that specifically binds to an antigen. In some embodiments, the recombinant receptor is a CAR that contains an extracellular antigen recognition domain that specifically binds to an antigen. In some embodiments, the ligand (e.g., antigen) is a protein expressed on the surface of a cell. In some embodiments, the CAR is a TCR-like CAR, and the antigen is a processed peptide antigen, such as a peptide antigen of an intracellular protein, that is recognized on the cell surface in the context of a Major Histocompatibility Complex (MHC) molecule, as with a TCR.
Exemplary recombinant receptors (including CARs and recombinant TCRs) and methods of engineering and introducing the receptor into cells include, for example, those described in the following documents: international patent application publication nos. WO 200014257, WO 2013126726, WO 2012/129514, WO 2014031687, WO 2013/166321, WO 2013/071154, WO 2013/123061, U.S. patent application publication nos. US2002131960, US2013287748, US20130149337, U.S. patent nos. 6,451,995, 7,446,190, 8,252,592, 8,339,645, 8,398,282, 7,446,179, 6,410,319, 7,070,995, 7,265,209, 7,354,762, 7,446,191, 8,324,353 and 8,479,118, and european patent application No. EP 2537416; and/or those described in the following documents: sadelain et al, cancer discover.2013, month 4; 3 (4) 388-398; davila et al (2013) PLoS ONE 8 (4): e61338; turtle et al, curr. Opin. Immunol, 10, 2012; 24 633-39; wu et al, cancer,2012, 3, 18 (2): 160-75. In some aspects, genetically engineered antigen receptors include CARs, such as those described in U.S. Pat. No. 7,446,190, and in international patent application publication No. WO/2014055668 A1.
In some embodiments, the engineered cells of the therapeutic cell composition contain a recombinant receptor (e.g., CAR) that binds to a tumor antigen. In some embodiments, the recombinant receptor specifically recognizes and/or targets an antigen associated with cancer and/or present on a universal label. In some embodiments of the present invention, in some embodiments, antigens recognized or targeted by recombinant receptors are B Cell Maturation Antigen (BCMA), ROR1, carbonic anhydrase 9 (CAIX), her2/neu (receptor tyrosine kinase erbB 2), L1-CAM, CD19, CD20, CD22, mesothelin, CEA and hepatitis B surface antigen, antifolate receptor, CD23, CD24, CD30, CD33, CD38, CD44, EGFR, epithelial glycoprotein 2 (EPG-2), epithelial glycoprotein 40 (EPG-40), EPHa2, erb-B3, erb-B4, erbB dimer, EGFRvIII, folate Binding Protein (FBP), FCRL5, FCRH5, fetal acetylcholine receptor, GD2, GD3, HMW-MAA, IL-22R-alpha, IL-13R-alpha 2, kinase insert domain receptor (kdr), IL-22R-alpha kappa light chain, lewis Y, L1 cell adhesion molecule (L1-CAM), melanomA-Associated antigen (MAGE) -A1, MAGE-A3, MAGE-A6, melanoma preferential expression antigen (PRAME), survivin, TAG72, B7-H6, IL-13 receptor alpha 2 (IL-13 Ra 2), CA9, GD3, HMW-MAA, CD171, G250/CAIX, HLA-AI MAGE Al, HLA-A2 NY-ESO-1, PSCA, folate receptor-a, CD44v6, CD44v7/8, avb6 integrin, 8H9, NCAM, VEGF receptor, 5T4, fetal AchR, NKG2D ligand, CD44v6, dual antigen, cancer-testis antigen, mesothelin, murine mucin 1 (MUC 1), MUC16, PSCA, NKG2D, NY-ESO-1, MART-1, gp100, carcinoembryonic antigen, ROR1, TAG72, VEGF-R2, carcinoembryonic antigen (CEA), her2/neu, estrogen receptor, progesterone receptor, ephrin B2, CD123, c-Met, GD-2, O-acetylated GD2 (OGD 2), CE7, wilms tumor 1 (WT-1), cyclin A2, CCL-1, CD138, human antigen optionally of any of the foregoing; antigens specific to pathogens. In some embodiments, the antigen recognized and/or targeted by the recombinant receptor is selected from the group consisting of: notch 1, notch 2, notch 3, notch4, cell surface associated mucin 1 (MUC 1), ephrin B2, betaglycan (TGFBR 3), CD43, CD44, CSF1R, CX CR1, CXCL16, δ1, E-cadherin, N-cadherin, HLA-A2, IFNaR2, IL1R1, IL1R2, IL6R, and Amyloid Precursor Protein (APP).
In some embodiments, the antigen recognized and/or targeted by the recombinant receptor is B Cell Maturation Antigen (BCMA). Exemplary antigen binding domains that target or specifically bind BCMA and CARs containing such antigen binding domains are known, see, e.g., WO 2016/090320, WO 2016090327, WO 2010104949A2, and WO 2017173256. In some embodiments, the antigen binding domain is an scFv comprising VH and VL derived from an antibody or antibody fragment specific for BCMA. In some embodiments, antibodies or antibody fragments that bind BCMA are or contain VH and VL from antibodies or antibody fragments described in international patent application publication nos. WO 2016/090327 and WO 2016/090320.
As described above, the assay may include a plurality of incubations, wherein each incubation is a culture of engineered cells containing a different stepwise adjustment ratio of the therapeutic cell composition with a recombinant receptor stimulator capable of stimulating recombinant receptors of the engineered cells to stimulate recombinant receptor-dependent activity (or vice versa). It is within the level of the skilled artisan to empirically determine the precise ranges or amounts of cells and receptor stimulators of the therapeutic cell composition to achieve a stepwise adjustment of the response in the assay. For example, the amount or quantity will depend on the particular form of the assay, such as the size of the vessel in which the assay is performed. It will be appreciated that the amount will be less when measured in a vessel having a smaller surface area than in a vessel having a larger surface area. Typically, the amount of cells is an amount in which the cells are sub-confluent (e.g., no more than 25% confluent or 50% confluent). In addition, the specific ratio ranges can be determined empirically based on the specific antigen and target cells employed. For example, the selected ratio is a ratio that includes a linear dose response increase in recombinant receptor-dependent activity across multiple stepwise adjustments of a reference standard. In some embodiments, the ratio is selected to further include a lower asymptote for receptor-dependent activity and a higher asymptote for receptor-dependent activity, which represent the minimum and maximum responses, respectively, of the reference standard.
In some embodiments, the number of engineered cells of the therapeutic cell composition is changed while the amount or concentration of binding molecules is kept constant to produce different ratios. In some embodiments, the number of cells of the therapeutic cell composition in each incubation is from at or about 1x10 4 Up to about 1x10 6 Individual cells, about 1x10 4 Up to about 9x10 5 Individual cells, about 1x10 4 Up to about 8x10 5 Individual cells, about 1x10 4 Up to about 7x10 5 Individual cells, about 1x10 4 Up to about 6x10 5 Individual cells, about 1x10 4 Up to about 5x10 5 Individual cells, about 1x10 4 Up to about 4x10 5 Individual cells, about 1x10 4 Up to about 3x10 5 Individual cells, about 1x10 4 Up to about 2x10 5 Individual cells, about 1x10 4 Up to about 1x10 5 Individual cells, about 1x10 4 Up to about 9x10 4 Individual cells, about 1x10 4 Up to about 8x10 4 Individual cells, about 1x10 4 Up to about 7x10 4 Individual cells, about 1x10 4 Up to about 6x10 4 Individual cells, about 1x10 4 Up to about 5x10 4 Individual cells, about 1x10 4 Up to about 4x10 4 Individual cells, about 1x10 4 Up to about 3x10 4 Individual cells, about 1x10 4 Up to about 2x10 4 Individual cells were gradually adjusted. In some embodiments, the cell number of the therapeutic cell composition is from at or about 1x10 in multiple incubations 4 Up to about 1x10 5 Individual cells, 1x10 4 Up to about 8x10 4 Individual cells, 1x10 4 Up to about 6x10 4 Individual cells, 1x10 4 Up to about 4x10 4 Individual cells, 1x10 4 Up to about 2x10 4 Individual cells were gradually adjusted. In some embodiments, the number of cells of the therapeutic cell composition is from at or about 10,000 to at or about 1,000,000 cells per cell over multiple incubationsAnd (5) step adjustment. In some embodiments, the cell number of the therapeutic cell composition is stepwise adjusted from at or about 10,000 to at or about 500,000 cells over multiple incubations. In some embodiments, the cell number of the therapeutic cell composition is stepwise adjusted from at or about 10,000 to at or about 250,000 cells over multiple incubations. In some embodiments, the cell number of the therapeutic cell composition is stepwise adjusted from at or about 10,000 to at or about 200,000 cells over multiple incubations. In some embodiments, the cell number of the therapeutic cell composition is stepwise adjusted from at or about 10,000 to at or about 150,000 cells over multiple incubations. In some embodiments, the cell number of the therapeutic cell composition is stepwise adjusted from at or about 10,000 to at or about 100,000 cells over multiple incubations. In some embodiments, the cell number of the therapeutic cell composition is stepwise adjusted from at or about 10,000 to at or about 50,000 cells over multiple incubations. In some embodiments, the cell number of the therapeutic cell composition in any of the foregoing is the total number of cells, the total number of living cells, the total number of car+ cells, the total number of cd8+ cells, the total number of cd4+ cells, the total number of cd3+ cells, the total number of cd8+/car+ cells, the total number of cd4+/car+ cells, or the total number of cd3+/car+ cells.
In some embodiments, the cell number of the therapeutic cell composition remains constant over multiple incubations, and the amount of recombinant receptor stimulant is gradually adjusted. In some embodiments, the constant number of cells of the therapeutic cell composition over multiple incubations is at about 1x10 4 Up to about 1x10 6 Individual cells, about 1x10 4 Up to about 9x10 5 Individual cells, about 1x10 4 Up to about 8x10 5 Individual cells, about 1x10 4 Up to about 7x10 5 Individual cells, about 1x10 4 Up to about 6x10 5 Individual cells, about 1x10 4 Up to about 5x10 5 Individual cells, about 1x10 4 Up to about 4x10 5 Individual cells, about 1x10 4 Up to about 3x10 5 Individual cells, about 1x10 4 Up to about 2x10 5 Individual cells, about 1x10 4 Up to about 1x10 5 Individual cells, about 1x10 4 Up to about 9x10 4 Individual cells, about 1x10 4 Up to about 8x10 4 Individual cells, about 1x10 4 Up to about 7x10 4 Individual cells, about 1x10 4 Up to about 6x10 4 Individual cells, about 1x10 4 Up to about 5x10 4 Individual cells, about 1x10 4 Up to about 4x10 4 Individual cells, about 1x10 4 Up to about 3x10 4 Individual cells, about 1x10 4 Up to about 2x10 4 Amount between individual cells. In some embodiments, the constant number of cells of the therapeutic cell composition over multiple incubations is from at or about 1x10 4 Up to about 1x10 5 Individual cells, 1x10 4 Up to about 8x10 4 Individual cells, 1x10 4 Up to about 6x10 4 Individual cells, 1x10 4 Up to about 4x10 4 Individual cells, 1x10 4 Up to about 2x10 4 Amount of individual cells. In some embodiments, the constant number of cells of the therapeutic cell composition over multiple incubations is an amount from or about 10,000 to or about 1,000,000 cells. In some embodiments, the constant number of cells of the therapeutic cell composition over multiple incubations is an amount from or about 10,000 to or about 500,000 cells. In some embodiments, the constant number of cells of the therapeutic cell composition over multiple incubations is an amount from or about 10,000 to or about 250,000 cells. In some embodiments, the constant number of cells of the therapeutic cell composition over multiple incubations is an amount from or about 10,000 to or about 150,000 cells. In some embodiments, the constant number of cells of the therapeutic cell composition over multiple incubations is an amount from or about 10,000 to or about 100,000 cells. In some embodiments, the constant number of cells of the therapeutic cell composition in each incubation in the plurality of incubations is an amount from or about 10,000 to or about 50,000 cells. In some embodiments, the constant number of cells of the therapeutic cell composition over multiple incubations is at or about 5,000, 10,000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, or 100,000 cells. In some embodiments, the constant number of cells of the therapeutic cell composition over multiple incubations is at or about 5,000, 10,000, 20,000, 30,000, 40,000, or 50,000 cells. In some embodiments of the present invention, in some embodiments, The constant number of cells of the therapeutic cell composition over multiple incubations is at or about 20,000, 30,000, 40,000, or 50,000 cells. In some embodiments, the constant number of cells of the therapeutic cell composition is at or about 50,000 cells over multiple incubations. In some embodiments, the constant number of cells of the therapeutic cell composition in any of the foregoing is the total number of cells, the total number of living cells, the total number of car+ cells, the total number of cd8+ cells, the total number of cd4+ cells, the total number of cd3+ cells, the total number of cd8+/car+ cells, the total number of cd4+/car+ cells, or the total number of cd3+/car+ cells.
B. Recombinant receptor stimulators
The methods of assessing potency provided herein include means for stimulating recombinant receptors (e.g., CAR, TCR) of engineered cells of a therapeutic cell composition. It is contemplated that any means suitable for stimulating recombinant receptors may be used that can also be quantified and delivered, such as cell to stimulation means to produce therapeutic cell compositions in different ratios. In some embodiments, the means for stimulating a recombinant receptor is achieved by a recombinant receptor stimulator capable of binding to the recombinant receptor and stimulating intracellular signaling through the recombinant receptor to produce recombinant receptor-dependent activity, as described in section I-C. Exemplary recombinant receptor stimulators include antigens of recombinant receptors (e.g., purified or recombinant antigens), antibodies (e.g., anti-idiotype antibodies), and antigen-expressing cells.
As described above, in some embodiments, multiple incubations of cells of therapeutic cell compositions with recombinant receptor stimulators at different stepwise adjustment ratios can be achieved by: a constant number of cells (e.g., live, car+, cd4+, cd8+, cd3+, cd4+/car+, cd8+/car+, cd3+/car+ cells) of a therapeutic cell composition are cultured with varying or stepwise adjusting amounts (e.g., concentration, mass) of recombinant receptor stimulators. In some embodiments, the amount or concentration of recombinant receptor stimulant is varied or stepwise adjusted by a factor of: is at or about 10,000 times, 5,000 times, 1,000 times, 1,500 times, 500 times, 250 times, 200 times, 150 times, 100 times, 75 times, 50 times, 25 times, or 10 times. In some embodiments, the amount or concentration of recombinant receptor stimulant is varied or stepwise adjusted by a factor of: between or about 10,000 to 100 times, 5,000 to 100 times, or 1,000 to 100 times. In some embodiments, the amount or concentration of recombinant receptor stimulant varies by a factor of: is at or about 5,000 times, 1,000 times, 1,500 times or 500 times. In some embodiments, the amount or concentration of recombinant receptor stimulant is varied or stepwise adjusted by a factor of: is at or about 5,000 times. In some embodiments, the amount or concentration of recombinant receptor stimulant is varied or stepwise adjusted by a factor of: is at or about 1,000 times. In some embodiments, the amount or concentration of recombinant receptor stimulant is varied or stepwise adjusted by a factor of: is at or about 500 times.
1. Immobilization of binding molecules and surfaces
In certain embodiments, the recombinant receptor stimulant is comprised of a binding molecule (or target) capable of being bound by a recombinant receptor. In some embodiments, the binding molecule is immobilized on a surface support. In provided embodiments, the binding molecule can be an antigen or portion of an antigen of a recombinant receptor (e.g., an extracellular portion of an antigen) or an antibody specific for a recombinant receptor (e.g., an anti-idiotype antibody). For example, a binding molecule (e.g., an antigen or binding portion thereof, or an anti-idiotype antibody) can be immobilized or bound to a surface support, such as a non-cellular particle, wherein a recombinant receptor-expressing cell (e.g., CAR-T cell) of a therapeutic composition, such as a stepwise-adjusting amount of the cell, is contacted with the surface support. In some embodiments, the particles (e.g., bead particles) described herein provide a solid support or matrix to which a binding molecule (e.g., an antigen or binding portion thereof, or an anti-idiotype antibody) can be bound or attached in a manner that allows for interaction between the binding molecule and a cell, particularly binding between the binding molecule and a recombinant receptor expressed on the cell surface (e.g., CAR). In certain embodiments, the interaction between the conjugated or attached binding molecule and the cell mediates stimulation of a recombinant receptor, including one or more recombinant receptor-dependent activities as described, such as activation, amplification, cytokine production, cytotoxic activity, or other activities, see, e.g., section i.c.
In certain embodiments, the surface support is a particle (e.g., a bead particle) immobilized or attached to a binding molecule (e.g., an antigen or binding portion thereof, or an anti-idiotype antibody). In some embodiments, the surface support is a solid support. In some examples, the solid support is a bead and the antigen or moiety is immobilized on the bead. In some embodiments, the solid support is a surface of a well or plate (e.g., a cell culture plate). In some embodiments, the surface support is a soluble oligomer particle and the antigen is immobilized on the surface of the soluble oligomer particle. Examples of surface supports for immobilization or attachment of agents (e.g. binding molecules) for recognition or binding of recombinant receptors can be found in published international application WO 2019/027850, which is incorporated by reference for all purposes.
In particular embodiments, the surface support is a particle, which may include a colloidal particle, microsphere, nanoparticle, bead (e.g., magnetic bead), or the like. In some embodiments, the particles or beads are biocompatible, i.e., non-toxic. In certain embodiments, the particles or beads are non-toxic to cultured cells (e.g., cultured T cells). In certain embodiments, the particles are monodisperse. In certain embodiments, "monodisperse" encompasses particles (e.g., bead particles) having a size dispersion that has a standard deviation of less than 5%, e.g., a diameter having a standard deviation of less than 5%.
In some embodiments, the particles or beads are biocompatible, i.e., are composed of a material suitable for biological use. In some embodiments, the particles (e.g., beads) are non-toxic to cultured cells (e.g., cultured T cells). In some embodiments, the particle (e.g., bead) may be any particle that is capable of attaching the binding molecule in a manner that allows for interaction between the binding molecule and the cell. In certain embodiments, the particles (e.g., beads) may be any particle that may be modified (e.g., surface functionalized) to allow attachment of binding molecules to the particle surface. In some embodiments, the particles (e.g., beads) are comprised of glass, silica, polyester of hydroxycarboxylic acid, polyanhydride of dicarboxylic acid, or copolymer of hydroxycarboxylic acid and dicarboxylic acid. In some embodiments, the particles (e.g., beads) may consist of, or at least consist of, the following: linear or branched, substituted or unsubstituted, saturated or unsaturated, linear or crosslinked alkyl (alkinyl), haloalkyl, thioalkyl, aminoalkyl, aryl, aralkyl, alkenyl, aralkenyl, heteroaryl, or polyesters of alkoxyhydroxy acids; or a linear or branched, substituted or unsubstituted, saturated or unsaturated, linear or crosslinked alkanyl, haloalkyl, thioalkyl, aminoalkyl, aryl, aralkyl, alkenyl, aralkenyl, heteroaryl or polyanhydride of an alkoxy dicarboxylic acid. Further, the particles (e.g., beads) may be, or consist of, quantum dots, such as quantum dot polystyrene particles (e.g., beads). Particles (e.g., beads) comprising a mixture of ester and anhydride linkages (e.g., copolymers of glycolic acid and sebacic acid) may also be employed. For example, the particles (e.g., beads) may comprise materials including: polyglycolic acid Polymer (PGA), polylactic acid Polymer (PLA), polysebacic acid Polymer (PSA), poly (lactic-co-glycolic acid) copolymer (PLGA), rho poly (lactic-co-sebacic acid) copolymer (PLSA), poly (glycolic acid-co-sebacic acid) copolymer (PGSA), and the like. Other polymers that may constitute the particles (e.g., beads) include caprolactone, carbonate, amide, amino acid, orthoester, acetal, cyanoacrylate, and degradable urethane polymers or copolymers, as well as copolymers of these with linear or branched, substituted or unsubstituted alkyl, haloalkyl, thioalkyl, aminoalkyl, alkenyl, or aromatic hydroxy acids or dicarboxylic acids. In addition, biologically important amino acids having reactive side chain groups (e.g., lysine, arginine, aspartic acid, glutamic acid, serine, threonine, tyrosine, and cysteine, or enantiomers thereof, may be included in copolymers having any of the above materials to provide reactive groups for conjugation with a binding molecule (e.g., a polypeptide antigen or antibody).
In some embodiments, the particles are beads having a diameter greater than 0.001 μm, greater than 0.01 μm, greater than 0.05 μm, greater than 0.1 μm, greater than 0.2 μm, greater than 0.3 μm, greater than 0.4 μm, greater than 0.5 μm, greater than 0.6 μm, greater than 0.7 μm, greater than 0.8 μm, greater than 0.9 μm, greater than 1 μm, greater than 2 μm, greater than 3 μm, greater than 4 μm, greater than 5 μm, greater than 6 μm, greater than 7 μm, greater than 8 μm, greater than 9 μm, greater than 10 μm, greater than 20 μm, greater than 30 μm, greater than 40 μm, greater than 50 μm, greater than 100 μm, greater than 500 μm, and/or greater than 1,000 μm. In some embodiments, the particles or beads have a diameter of between or about 0.001 μm and 1,000 μm, between 0.01 μm and 100 μm, between 0.1 μm and 10 μm, between 0.1 μm and 100 μm, between 0.1 μm and 10 μm, between 0.001 μm and 0.01 μm, between 0.01 μm and 0.1 μm, between 0.1 μm and 1 μm, between 1 μm and 10 μm, between 1 μm and 2 μm, between 2 μm and 3 μm, between 3 μm and 4 μm, between 4 μm and 5 μm, between 1 μm and 5 μm, and/or between 5 μm and 10 μm (each inclusive). In certain embodiments, the particles or beads have an average diameter of 1 μm and 10 μm (each inclusive). In certain embodiments, the particles (e.g., beads) have a diameter of at or about 1 μm. In particular embodiments, the particles (e.g., beads) have an average diameter of at or about 2.8 μm. In some embodiments, the particles (e.g., beads) have a diameter of at or about 4.8 μm.
Particles (e.g., bead particles) for use in the methods described herein may be produced or commercially obtained. Particles (e.g., beads), including methods of producing particles (e.g., beads), are well known in the art. See, for example, U.S. patent No. 6,074,884;5,834,121;5,395,688;5,356,713;5,318,797;5,283,079;5,232,782;5,091,206;4,774,265;4,654,267;4,554,088;4,490,436;4,452,773; U.S. patent application publication No. 20100207051; and sharp, pau t., methods of Cell Separation, elsevier,1988. Commercially available particles (e.g., beads) (e.g., bead particles) include, but are not limited to, proMagTM (Polysciences, inc.); comp ltm (PolySciences, inc.);(PolySciencs, inc.), comprising +.>Plus (Polysciences, inc.) and +.>Maxi(Bang Laboratories,Inc.);M-PVA(Cehmagen Biopolymer Technologie AG);SiMAG(Chemicell GmbH);beadMAG(Chemicell GmbH);(Cortex Biochem);/>(Invitrogen), comprising->M-280 sheep anti-rabbit IgG (Invitrogen), < >>FlowCompTM (e.g.)>FlowCompTMHuman CD3,Invitrogen)、/>M-450 (e.g., ->M-450Tosylactivated,Invitrogen)、/>UntuchedTM (e.g.)>Untouchem human CD 8T cells, invitrogen), and +.>(e.g., for T cell expansion and activation)Human T-activator CD3/CD28, invitrogen); />M(Merk Chimie SAS);EM (Merk Chimie SAS); macsibeads (tm) particles (e.g., avidin MACSiBead particles, miltenyi Biotec, catalog No. 130-091-147); / >Magnetic beads (IBA biotag technology);magnetic beads (IBA biotag technology); />(Micormod Partikeltechnologie GmbH)/>(Micromod Partikeltechnologie);MagneSilTM(Promega GmbH);MGP(Roche Applied Science Inc.);Pierce TM Protein G magnetic beads (Thermo Fisher Scientific inc.); pierce TM Protein a magnetic beads (Thermo Fisher Scientific inc.); pierce TM Protein a/G magnetic beads (Thermo Fisher Scientific inc.); pierce TM NHS activated magnetic beads (Thermo Fisher Scientific inc.); pierce TM Protein L magnetic beads (Thermo Fisher Scientific inc.); pierce TM anti-HA magnetic beads (Thermo Fisher Scientific inc.); pierce TM anti-c-Myc magnetic beads (Thermo Fisher Scientific inc.); pierce TM Glutathione magnetic beads (Thermo Fisher Scientific inc.); pierce TM Streptavidin magnetic beads (Thermo Fisher Scientific inc.); magnaBindTM magnetic beads (Thermo Fisher Scientific inc.); sera-mag (TM) magnetic beads (Thermo Fisher Scientific Inc.); anti->M2 magnetic beads (Sigma-Aldrich); spharotm magnetic particles (Spherotech inc.); and HisPurTM Ni-NTA magnetic beads (Thermo Fisher Scientific inc.).
In certain embodiments, the antigen or extracellular domain portion thereof is bound to a particle (e.g., a bead) via a covalent chemical bond. In certain embodiments, the reactive group or moiety of an amino acid of an antigen or extracellular domain portion thereof is conjugated directly to the reactive group or moiety on the particle surface by a direct chemical reaction. In certain embodiments, the amino acid carboxyl group (e.g., C-terminal carboxyl group), hydroxyl group, thiol, or amine group (e.g., amino acid side chain group) of the antigen or extracellular binding portion thereof is conjugated directly to the hydroxyl or carboxyl group of the PLA or PGA polymer, the terminal amine or carboxyl group of the dendrimer, or the hydroxyl, carboxyl, or phosphate group of the phospholipid on the particle surface by a direct chemical reaction. In some embodiments, the conjugate moiety is conjugated (e.g., covalently bound) to both the binding molecule and the particle, thereby linking them together.
In certain embodiments, the surface of the particle comprises chemical moieties and/or functional groups that allow for the attachment (e.g., covalent, non-covalent) of binding molecules (e.g., polypeptide antigens or antibodies). In certain embodiments, the particle surface contains exposed functional groups. Suitable surface-exposed functional groups include, but are not limited to, carboxyl, amino, hydroxyl, sulfuric acid, tosyl, epoxy, and chloromethyl groups. In some embodiments, the binding molecule is a polypeptide and is conjugated to a surface-exposed functional group. In some embodiments, the surface-exposed functional group must be activated, i.e., it must undergo a chemical reaction to produce an intermediate product capable of directly binding the polypeptide. For example, the carboxyl groups of the polypeptide molecules may be activated with the agents described above to produce intermediate esters capable of direct binding to the surface-exposed amino groups of the particles. In other examples, sulfosuccinimidyl (4-iodoacetyl) aminobenzoate (sulfo-SIAB) chemistry can be used to covalently bind free amine groups on the surface of a surface support (e.g., a bead) to an antigenic peptide and protein, or an antigenic peptide or protein fusion protein. In still other particular embodiments, the polypeptide binding molecule is covalently attached to the particle (e.g., bead particle) at a surface exposed functional group that does not require activation by an agent prior to formation of the covalent attachment. Examples of such functional groups include, but are not limited to, tosyl, epoxy, and chloromethyl.
In some embodiments, the antigen may be conjugated to a support (e.g., a bead) by a non-covalent bond between a ligand that binds to the antigen peptide or protein and an anti-ligand attached to the surface support (e.g., a bead). In some embodiments, a biotin ligase recognition sequence tag may be attached to the C-terminus of an antigenic peptide or protein, and such tag may be biotinylated by the biotin ligase. Biotin may then be used as a ligand to non-covalently conjugate the antigenic peptide or protein with avidin or streptavidin that is adsorbed or otherwise bound to the support surface as an anti-ligand. Alternatively, if a binding molecule (e.g., antigen) is fused to an immunoglobulin domain bearing an Fc region as described herein, the Fc domain may act as a ligand and protein a, covalently or non-covalently bound to the surface of a surface support (e.g., bead), may be used as an anti-ligand to non-covalently conjugate an antigenic peptide or protein to a carrier. Other means that can be used to non-covalently conjugate a binding molecule (e.g., an antigen or an anti-idiotype antibody) to a surface support (e.g., a bead) are well known in the art, including metal ion chelation techniques (e.g., using a polyHis tag at the C-terminus of the binding molecule (e.g., an antigen) and Ni-coated surface supports), and these methods can be substituted for those described herein.
In some embodiments, a binding molecule (e.g., an antigen or an anti-idiotype antibody) is conjugated to the particle through a linker. In certain embodiments, the linker may include, but is not limited to, a variety of bifunctional protein coupling agents such as N-succinimidyl-3- (2-pyridyldithio) propionate (SPDP), succinimidyl-4- (N-maleimidomethyl) cyclohexane-l-carboxylate, iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyldiimidinate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutaraldehyde), bis-azido compounds (such as bis (p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis- (p-diazoniumbenzoyl) -ethylenediamine), diisocyanates (such as toluene 2, 6-diisocyanate), and bis-active fluorine compounds (such as l, 5-difluoro-2, 4-dinitrobenzene). Specific coupling agents include N-succinimidyl-3- (2-pyridyldithio) propionate (SPDP) and N-succinimidyl-4- (2-pyridylthio) valerate (SPP) to provide disulfide bonds.
a. Antigens
In some embodiments, the recombinant receptor stimulant is or includes an antigen, such as a recombinant antigen or fragment thereof. For example, the recombinant receptor stimulant can be an antigen immobilized or bound to a surface support, such as a microplate, a solid particle (e.g., a bead), or an oligomer particle, e.g., as described above. In some embodiments, the antigen is a polypeptide or variant or fragment of a polypeptide expressed on the surface of a cell associated with a disease (e.g., a cancer cell and/or a tumor cell). It is understood that an antigen is an antigen that is recognized or bound by the extracellular domain of a recombinant receptor. The skilled artisan can determine antigens and antigen forms (e.g., cells expressed or immobilized on a solid surface) sufficient to stimulate a recombinant receptor.
In some embodiments, the antigen is or comprises αvβ6 integrin (avb 6 integrin), B Cell Maturation Antigen (BCMA), B7-H3, B7-H6, carbonic anhydrase 9 (CA 9, also known as CAIX or G250), cancer-testis antigen, cancer/testis antigen 1B (CTAG, also known as NY-ESO-1 and rage-2), carcinoembryonic antigen (CEA), cyclin A2, C-C motif chemokine ligand 1 (CCL-1), CD19, CD20, CD22, CD23, CD24, CD30, CD33, CD38, CD44v6, CD44v7/8, CD123, CD133, CD138, CD171, chondroitin sulfate proteoglycan 4 (CSPG 4), epidermal growth factor protein (EGFR), epidermal growth factor receptor type III mutant (EGFR vIII), epithelial glycoprotein 2 (EPG-2), epithelial glycoprotein 40 (EPG-40), hepadin B2, liver hormone receptor A2 (fcfc receptor 5), and the like receptor 5; also known as Fc receptor homolog 5 or FCRH 5), fetal acetylcholine receptor (fetal AchR), folic acid binding protein (FBP), folic acid receptor alpha, ganglioside GD2, O-acetylated GD2 (OGD 2), ganglioside GD3, glycoprotein 100 (gp 100), glypican-3 (GPC 3), G-protein coupled receptor 5D (GPRC 5D), her2/neu (receptor tyrosine kinase erb-B2), her3 (erb-B3), her4 (erb-B4), erbB dimer, human high molecular weight melanomA-Associated antigen (HMW-MAA), hepatitis B surface antigen, human leukocyte antigen A1 (HLa-A1), human leukocyte antigen A2 (HLa-A2), IL-22 receptor alpha (IL-22 ra), IL-13 receptor alpha 2 (IL-13 ra 2), kinase insert domain receptor (kdr), kappa light chain, L1 cell adhesion molecule (L1-CAM), CE7 epitope of L1-CAM, protein 8 family member a containing leucine rich repeat (LRRC 8A), lewis Y, melanomA-Associated antigen (MAGE) -A1, MAGE-A3, MAGE-A6, MAGE-a10, mesothelin (MSLN), c-Met, murine cytomegalovirus (MUC 1), MUC16, natural killer cell group 2 member D (g 2D) ligand, melanoma (MART 1), human cell adhesion molecule (nct 1), human prostate specific antigen (TRP), protein-associated protein (TRP 1), protein-specific receptor (TRP 1, p 2), protein-associated tumor antigen (TRP 1, p-specific tumor antigen (TRP 1), protein receptor-associated protein 2 (TRP 1), tumor-associated tumor antigen (TRP 1, tumor antigen (tumor antigen-specific tumor antigen), projection 1, tumor antigen (TRP 1, tumor antigen (tumor antigen), projection 1, tumor antigen (tumor antigen), tumor antigen (tumor antigen) protein 2), also known as dopachrome tautomerase, dopachrome delta isomerase, or DCT), vascular Endothelial Growth Factor Receptor (VEGFR), vascular endothelial growth factor receptor 2 (VEGFR 2), nephroblastoma 1 (WT-1), pathogen-specific or pathogen-expressed antigen, or antigen associated with a universal tag, and/or biotinylated molecules, and/or molecules expressed by HIV, HCV, HBV or other pathogens. In some embodiments, the receptor-targeted antigen comprises an antigen associated with a B cell malignancy, such as any of a number of known B cell markers. In some embodiments, the antigen is or comprises CD20, CD19, CD22, ROR1, CD45, CD21, CD5, CD33, igκ, igλ, CD79a, CD79b, or CD30.
In some embodiments, the antigen is or comprises a portion of a polypeptide antigen that is recognized or bound by a recombinant receptor (e.g., CAR). In certain embodiments, the portion of the antigen is a region containing an epitope recognized or bound by a recombinant receptor (e.g., CAR). In certain embodiments, the portion of the polypeptide antigen comprises about or at least 10, 15, 20, 25, 30, 35, 40, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 250, 300, 400, or 500 amino acids (in some cases consecutive amino acids) of the polypeptide recognized or bound by the recombinant receptor and or CAR. In certain embodiments, the polypeptide portion comprises an amino acid sequence of an epitope recognized by the recombinant receptor and/or CAR.
In certain embodiments, the antigen or moiety is a polypeptide variant that contains about or at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99%, or 99.5% amino acid sequence identity to a polypeptide that binds to and/or is recognized by a recombinant receptor and/or CAR.
In certain embodiments, the extracellular domain of a recombinant receptor (e.g., CAR) is specific for or binds to BCMA, and the antigen is BCMA or an extracellular domain portion of BCMA. In some embodiments, the BCMA polypeptide is a mammalian BCMA polypeptide. In a particular embodiment, the BCMA polypeptide is a human BCMA polypeptide. In some embodiments, the BCMA antigen is or comprises an extracellular domain of BCMA or portion thereof comprising an epitope recognized by an antigen receptor (e.g., CAR). In certain embodiments, a BCMA antigen is or comprises a polypeptide having an amino acid sequence having at least 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID No. 13, or a fragment of said polypeptide comprising at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 170, or at least 180 consecutive amino acids of SEQ ID No. 13. In some embodiments, the BCMA antigen is or includes the sequence shown in SEQ ID NO. 13, or a portion thereof, which is or contains an epitope recognized by an antigen receptor (e.g., a CAR).
In certain embodiments, the extracellular domain of a recombinant receptor (e.g., CAR) is specific for ROR1 or binds to ROR1, and the antigen is ROR1 or is an extracellular domain portion of ROR 1. In certain embodiments, the ROR1 polypeptide is mammalian. In certain embodiments, the ROR1 polypeptide is human. In some embodiments, the antigen is an extracellular domain of ROR1, or a portion thereof, comprising an epitope recognized by an antigen receptor (e.g., CAR). In some embodiments, the antigen is a polypeptide having an amino acid sequence with at least 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID No. 19, or a fragment of said polypeptide containing at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 170, or at least 180 consecutive amino acids of SEQ ID No. 19. In some embodiments, the ROR1 antigen comprises the sequence shown in SEQ ID NO. 19, or a portion thereof, comprising an epitope recognized by an antigen receptor (e.g., a CAR).
In certain embodiments, the extracellular domain of a recombinant receptor (e.g., CAR) is specific for CD22 or binds to CD22, and the antigen is CD22 or an extracellular domain portion of CD 22. In certain embodiments, the CD22 polypeptide is mammalian. In certain embodiments, the CD22 polypeptide is human. In some embodiments, the antigen is an extracellular domain of CD22, or a portion thereof, that comprises an epitope recognized by an antigen receptor (e.g., CAR). In some embodiments, the antigen is a polypeptide having an amino acid sequence with at least 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID No. 14, or a fragment of said polypeptide containing at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 170, or at least 180 consecutive amino acids of SEQ ID No. 14. In some embodiments, the CD22 antigen comprises the sequence shown in SEQ ID NO. 14, or a portion thereof, comprising an epitope recognized by an antigen receptor (e.g., a CAR).
In certain embodiments, the extracellular domain of a recombinant receptor (e.g., CAR) is specific for CD19 or binds to CD19, and the antigen is CD19 or an extracellular domain portion of CD 19. In certain embodiments, the CD19 polypeptide is mammalian. In certain embodiments, the CD19 polypeptide is human. In some embodiments, the antigen is an extracellular domain of CD19, or a portion thereof, that comprises an epitope recognized by an antigen receptor (e.g., CAR). In some embodiments, the antigen is a polypeptide having an amino acid sequence with at least 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID No. 15, or a fragment of said polypeptide containing at least 50, at least 55, at least 60, at least 65, at least 70, at least 75, at least 80, at least 85, at least 90, at least 95, at least 100, at least 110, at least 120, at least 130, at least 140, at least 150, at least 160, at least 170, or at least 180 consecutive amino acids of SEQ ID No. 15. In some embodiments, the CD19 antigen comprises the sequence shown in SEQ ID NO. 15, or a portion thereof, comprising an epitope recognized by an antigen receptor (e.g., a CAR).
In some embodiments, the antigen or portion thereof may be formed as a multimer (e.g., dimer) comprising two or more polypeptide antigens or portions or variants thereof that are recognized and/or bound by a recombinant receptor, such as an antigen receptor (e.g., CAR). In some embodiments, the polypeptide antigens or portions thereof are identical. In certain embodiments, the polypeptide antigens are directly or indirectly linked to a region or domain (e.g., a multimerization domain) that facilitates or stabilizes interactions between two or more polypeptide antigens by complementary interactions between the domains or regions. In some embodiments, providing the polypeptide antigen as a multimer (e.g., a dimer) provides multivalent interactions between the antigen or extracellular domain portion thereof and the antigen binding domain of an antigen receptor (e.g., CAR), which in some aspects can increase the avidity of the interaction. In some embodiments, increased avidity may be beneficial for stimulating or agonist activity of an antigen receptor (e.g., CAR) by an antigen or extracellular domain portion thereof conjugated to a bead.
In some embodiments, the polypeptide is directly or indirectly linked to a multimerization domain. Exemplary multimerization domains include immunoglobulin sequences or portions thereof, leucine zippers, hydrophobic regions, hydrophilic regions, and compatible inter-protein interaction domains. For example, the multimerization domain may be an immunoglobulin constant region or domain, such as, for example, an Fc domain from IgG (including IgG1, igG2, igG3, or IgG4 subtypes), igA, igE, igD, and IgM, and modified versions thereof, or portions thereof. In certain embodiments, the polypeptide antigen is directly or indirectly linked to an Fc domain. In some embodiments, the polypeptide is a fusion polypeptide comprising a polypeptide antigen or portion thereof and an Fc domain.
In certain embodiments, the antigen or extracellular domain portion thereof is a fusion polypeptide comprising an Fc domain. In some embodiments, the Fc domain consists of the second and third constant domains (i.e., CH2 and CH3 domains) of a heavy chain of the IgG, igA or IgD isotype (e.g., igG. CH2 or CH 3) of IgA and IgD isotypes. In some embodiments, the Fc domain consists of three heavy chain constant domains (i.e., CH2, CH3, and CH4 domains) of IgM or IgE isotype. In some embodiments, the Fc domain may further comprise a hinge sequence or portion thereof. In certain aspects, the Fc domain comprises part or all of the hinge domain and the CH2 and CH3 domains of an immunoglobulin molecule. In some cases, an Fc domain may form a dimer of two polypeptide chains joined by one or more disulfide bonds. In some embodiments, the Fc domain is derived from an immunoglobulin (e.g., igG, igA, igM or IgE) of a suitable mammal (e.g., human, mouse, rat, goat, sheep, or monkey). In some embodiments, the Fc domain comprises IgG C H 2 and C H 3 domain. In certain embodiments, the Fc domain is fused to the C-terminus of the polypeptide antigen. In certain embodiments, the Fc domain is fused to the N-terminus of the polypeptide antigen.
In some embodiments, the Fc domain is an IgG Fc domain or a portion or variant thereof. In some embodiments, the Fc domain is a human IgG Fc domain or a portion or variant thereof comprising the amino acid sequence set forth in SEQ ID No. 16, or an amino acid sequence having at least 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence identity to the sequence set forth in SEQ ID No. 16. In particular embodiments, the Fc domain is a wild-type human IgG Fc domain or a portion or variant thereof. In certain embodiments, the Fc domain is a variant of a wild-type human IgG1 Fc domain.
In some embodiments, the fusion polypeptide comprises a variant Fc domain. In certain embodiments, the variant human IgG Fc domain contains mutations (e.g., substitutions, deletions, or insertions) that reduce, and/or detract from pairing between the Fc domain and the light chain. In some embodiments, the variant human IgG Fc domain contains mutations that reduce the binding affinity between the Fc domain and the Fc receptor. In particular embodiments, the variant human IgG Fc domain contains mutations that reduce, and/or detract from the probability or likelihood of interaction between the Fc domain and the Fc receptor. In some embodiments, the variant human IgG Fc domain contains mutations that reduce the binding affinity between the Fc domain and a protein of the complement system. In particular embodiments, the variant human IgG Fc domain contains mutations that reduce, and/or detract from the probability or likelihood of interaction between the Fc domain and a protein of the complement system.
In some embodiments, the antigen or portion thereof is linked to a variant human IgG1 Fc domain. In some embodiments, the variant human IgG Fc domain contains a cystine to serine substitution in the hinge region of the Fc domain. In some embodiments, the variant human IgG Fc domain contains a leucine to alanine substitution in the hinge region of the Fc domain. In certain embodiments, the variant human IgG Fc domain contains a glycine to alanine substitution in the hinge region. In certain embodiments, the variant human IgG Fc domain contains alanine to serine substitutions in the CH2 region of the Fc domain. In some embodiments, the variant human IgG Fc domain comprises a proline to serine substitution in the CH2 region of the Fc domain. In some embodiments, the variant human IgG Fc domain comprises the amino acid sequence shown as SEQ ID NO. 17.
In some embodiments, the antigen or extracellular domain portion thereof is provided as a fusion polypeptide comprising an Fc domain, wherein the Fc domain is present at the C-terminus of the fusion polypeptide.
In some embodiments, the antigen and multimerization domains (e.g., fc domains) are linked by a linker (e.g., an amino acid linker). In certain embodiments, the antigen is fused to the N-terminus of the amino acid linker, and the multimerization domain (e.g., fc domain) is fused to the C-terminus of the linker. Although the amino acid linker may be of any length and contain any combination of amino acids, the linker length may be relatively short (e.g., ten or fewer amino acids) to reduce interactions between the linked domains. The amino acid composition of the linker may also be adjusted to reduce the number of amino acids with large side chains or amino acids that may be incorporated into secondary structures. Suitable amino acid linkers include, but are not limited to, those up to 3, 4, 5, 6, 7, 10, 15, 20, or 25 amino acids in length. Representative amino acid linker sequences include GGGGS (SEQ ID NO: 22), and linkers comprising 2, 3, 4, or 5 copies of GGGGS (SEQ ID NO: 22).
In some embodiments, the antigen is provided as an extracellular domain of BCMA (e.g., human BCMA) (BCMA-Fc) fused to an Fc domain. In particular embodiments, the BCMA-Fc antigen contains all or part of the amino acid sequence shown in SEQ ID No. 18, or an amino acid sequence that exhibits at least 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID No. 18 and comprises an epitope recognized by an antigen receptor (e.g., CAR).
In some embodiments, the antigen is provided as an extracellular domain of ROR1 (e.g., human ROR 1) (ROR 1-Fc) fused to an Fc domain. In certain embodiments, the ROR-1-Fc antigen contains all or part of the amino acid sequence shown in SEQ ID No. 20, or an amino acid sequence that exhibits at least 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID No. 20 and comprises an epitope recognized by an antigen receptor (e.g., CAR).
In particular embodiments, the antigen is provided as an extracellular domain of CD22 (e.g., human CD 22) (CD 22-Fc) fused to an Fc domain. In certain embodiments, the CD22-Fc antigen contains all or part of the amino acid sequence shown in SEQ ID No. 21, or an amino acid sequence that exhibits at least 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% sequence identity to SEQ ID No. 21 and comprises an epitope recognized by an antigen receptor (e.g., CAR).
In some embodiments, the Fc fusion of an antigen or extracellular binding domain thereof is linked or attached to a surface support as a dimer formed from two Fc fusion polypeptides comprising a polypeptide antigen or portion thereof and an Fc domain. In some embodiments, the resulting polypeptide antigen-Fc fusion protein (e.g., BCMA-Fc, ROR1-Fc, CD22-Fc, or CD 19-Fc) can be expressed in, for example, host cells transformed with an expression vector, such that assembly between Fc domains can occur through interchain disulfide bonds formed between Fc portions to produce a dimeric (e.g., bivalent) polypeptide antigen fusion protein. In some embodiments, the host cell is a mammalian cell line. Examples of mammalian cells for recombinant expression of proteins include HEK293 cells or CHO cells or derivatives thereof. In some aspects, the nucleic acid encoding the Fc fusion protein further comprises a signal peptide for secretion from a cell. In an exemplary embodiment, the signal peptide is CD33 (e.g., as shown in SEQ ID NO: 12).
In some embodiments, cells of the therapeutic cell composition express a CAR that binds to or recognizes a universal tag that can be fused to an antibody or fragment or variant thereof. In certain embodiments, cells expressing such CARs are capable of specifically recognizing and killing target cells (e.g., tumor cells) that have been bound by antibodies fused to a universal tag. One example includes, but is not limited to, anti-FITC CAR-expressing T cells that can bind to and/or recognize various human cancer cells when those cells are bound by a cancer-reactive FITC-labeled antibody. Thus, in some embodiments, the same CAR bound to a universal tag can be used to treat different cancers, provided that there is an available antibody containing a universal tag that recognizes a cancer-associated antigen. In certain embodiments, the particle (e.g., bead particle) comprises a surface exposed binding molecule comprising a universal tag binding molecule capable of binding or recognizing by a recombinant receptor (e.g., CAR). In certain embodiments, the binding molecule is a universal tag or portion thereof that is bound or recognized by an antigen receptor (e.g., CAR). Particular embodiments contemplate that any polypeptide domain that may be fused to an antibody or antigen-binding fragment or variant thereof that does not prevent binding of the antibody to its respective target is suitable for use as a universal tag. In some embodiments, the particle is bound to a binding molecule comprising a universal tag or portion thereof selected from the group consisting of: FITC, streptavidin, biotin, histidine, dinitrophenol, polymethylchlorophyll protein complex, green fluorescent protein, PE, HRP, palmitoylation, nitrosylation, alkaline phosphatase (alkalanine phosphatase), glucose oxidase, and maltose binding protein.
b. Antibodies to
In some aspects, the binding molecule is an antibody or antigen-binding fragment thereof that specifically recognizes a recombinant receptor (e.g., CAR). In some embodiments of these aspects, the antibody or antigen binding fragment specifically recognizes (e.g., specifically binds to an epitope on) an extracellular portion of a recombinant receptor (e.g., CAR).
In some aspects, the binding molecule is an anti-idiotype antibody or antigen-binding fragment thereof ("anti-ID") that specifically recognizes a recombinant receptor (e.g., a recombinant receptor, such as a CAR, as described in section III). In particular, the anti-idiotype antibody targets an antigen binding site of another antibody, such as an scFv of the extracellular antigen binding domain of the CAR. In some embodiments, the anti-ID is capable of binding to a recombinant receptor to stimulate recombinant receptor-dependent activity. Exemplary anti-idiotype antibodies to antigen-specific CARs are known. These include, but are not limited to, anti-idiotype antibodies against CD22 directed CARs, see, e.g., PCT publication No. WO 2013188864; anti-idiotype antibodies against CD19 directed CARs, see, e.g., PCT publication No. WO 2018/023100; anti-idiotype antibodies against GPRC5D directed CARs, see, e.g., PCT application No. PCT/US 2020/06397; and anti-idiotype antibodies against BCMA-directed CARs, see, e.g., PCT application No. PCT/US 2020/063292. As described above, the anti-idiotype antibody may be immobilized or attached to a surface support (e.g., a bead) to act as a recombinant receptor stimulator against cells expressing a recombinant receptor (e.g., CAR) targeted by the anti-idiotype antibody.
The term "antibody" is used herein in the broadest sense and includes polyclonal and monoclonal antibodies, including whole antibodies and functional (antigen-binding) antibody fragments, including antigen-binding fragment (Fab) fragments, F (ab') 2 Fragments, fab' fragments, fv fragments, recombinant IgG (rIgG) fragments, single chainAntibody fragments (including single chain variable fragments (scFv)) and single domain antibody (e.g., sdAb, sdFv, nanobody) fragments. The term encompasses genetically engineered and/or otherwise modified forms of immunoglobulins, such as intracellular antibodies, peptibodies, chimeric antibodies, fully human antibodies, humanized antibodies and heteroconjugated antibodies, multispecific (e.g., bispecific) antibodies, diabodies, triabodies and tetrabodies, tandem di-scFv, tandem tri-scFv. Unless otherwise indicated, the term "antibody" should be understood to encompass functional antibody fragments thereof. The term also encompasses whole or full length antibodies, including antibodies of any class or subclass (including IgG and subclasses thereof, igM, igE, igA and IgD).
The term "anti-idiotype antibody" refers to an antibody (including antigen-binding fragments thereof) that specifically recognizes, specifically targets, and/or specifically binds to a unique site (e.g., antigen-binding fragment) of the antibody. Unique positions of an antibody may include, but are not necessarily limited to, residues within one or more Complementarity Determining Regions (CDRs) of the antibody, variable regions of the antibody, and/or portions of such variable regions and/or such CDRs, and/or any combination of the foregoing. The CDRs may be one or more selected from the group consisting of: CDR-H1, CDR-H2, CDR-H3, CDR-L1, CDR-L2 and CDR-L3. The variable region of an antibody may be a heavy chain variable region, a light chain variable region, or a combination of a heavy chain variable region and a light chain variable region. A fragment or portion of a heavy chain variable region and/or a light chain variable region of an antibody may be a fragment comprising 2 or more, 5 or more, or 10 or more contiguous amino acids within the heavy chain variable region or the light chain variable region of the antibody, for example from about 2 to about 100, from about 5 to about 100, from about 10 to about 100, from about 2 to about 50, from about 5 to about 50, or from about 10 to about 50 contiguous amino acids; the unique positions may include a plurality of discrete stretches of amino acids. A partial fragment of the heavy and light chain variable regions of an antibody may be a fragment comprising 2 or more, 5 or more, or 10 or more contiguous amino acids within the variable region, for example from about 2 to about 100, from about 5 to about 100, from about 10 to about 100, from about 2 to about 50, from about 5 to about 50, or from about 10 to about 50 contiguous amino acids, and in some embodiments, containing one or more CDRs or CDR fragments. CDR fragments may be 2 or more, or 5 or more amino acids, contiguous or non-contiguous within a CDR. Thus, a unique position of an antibody may be from about 2 to about 100, from about 5 to about 100, from about 10 to about 100, from about 2 to about 50, from about 5 to about 50, or from about 10 to about 50 consecutive amino acids within the heavy chain variable region or the light chain variable region of the antibody that contain one or more CDRs or one or more CDR fragments. In another embodiment, the unique position may be a single amino acid located in a variable region (e.g., CDR site) of an antibody.
In some embodiments, the unique position is any single epitope or epitope within the variable portion of the antibody. In some cases, it may overlap with the actual antigen binding site of the antibody, and in some cases, it may comprise a variable region sequence outside the antigen binding site of the antibody. In some embodiments, a collection of individual unique bits of an antibody is referred to as the "idiotype" of such an antibody.
The terms "complementarity determining region" and "CDR" are synonymous with "hypervariable region" or "HVR," and are known in the art to refer to non-contiguous amino acid sequences within the variable region of an antibody that confer antigen specificity and/or binding affinity. Typically, there are three CDRs (CDR-H1, CDR-H2, CDR-H3) in each heavy chain variable region, and three CDRs (CDR-L1, CDR-L2, CDR-L3) in each light chain variable region. "framework region" and "FR" are known in the art to refer to the non-CDR portions of the variable regions of the heavy and light chains. Typically, there are four FRs (FR-H1, FR-H2, FR-H3 and FR-H4) in each full-length heavy chain variable region, and four FRs (FR-L1, FR-L2, FR-L3 and FR-L4) in each full-length light chain variable region.
The exact amino acid sequence boundaries for a given CDR or FR can be readily determined using any of a number of well known schemes, including those described in the following documents: kabat et al (1991), "Sequences of Proteins of Immunological Interest," 5 th edition Public Health Service, national Institutes of Health, bethesda, MD ("Kabat" numbering scheme); al-Lazikani et Al, (1997) JMB 273,927-948 ("Chothia" numbering scheme); macCallum et al, J.mol. Biol.262:732-745 (1996), "anti-body-antigen interactions: contact analysis and binding site topography," J.mol. Biol.262,732-745 "(" Contact "numbering scheme); lefranc MP et al, "IMGT unique numbering for immunoglobulin and T cell receptor variabledomains and Ig superfamily V-like domains," Dev Comp Immunol, month 1 2003; 27 (1) 55-77 ("IMGT" numbering scheme); and Honyger A and Pluckthun A, "Yet another numbering scheme for immunoglobulin variabledomains: an automatic modeling and analysis tool," J Mol Biol, 6/8/2001; 309 (3) 657-70 ("Aho" numbering scheme).
The boundaries of a given CDR or FR may vary depending on the scheme used for identification. For example, the Kabat scheme is based on structural alignment, while the Chothia scheme is based on structural information. Numbering of the Kabat and Chothia protocols is based on the most common antibody region sequence length, with insertions provided by insert letters such as "30a" and deletions in some antibodies. Both of these schemes place certain insertions and deletions ("indels") at different positions, resulting in different numbers. The Contact scheme is based on analysis of complex crystal structures and is similar in many respects to the Chothia numbering scheme.
Table 1 below lists exemplary location boundaries for CDR-L1, CDR-L2, CDR-L3, and CDR-H1, CDR-H2, CDR-H3 identified by the Kabat, chothia and Contact schemes, respectively. For CDR-H1, residue numbers are listed using the two numbering schemes of Kabat and Chothia. FR is located between CDRs, e.g., FR-L1 is located between CDR-L1 and CDR-L2, and so on. It should be noted that because the Kabat numbering scheme shown places insertions at H35A and H35B, when numbered using the Kabat numbering convention shown, the ends of the Chothia CDR-H1 loop vary between H32 and H34 depending on the length of the loop.
Table 1 CDR boundaries according to various numbering schemes.
1-Kabat et al (1991), "Sequences of Proteins of Immunological Interest," 5 th edition Public Health Service, national Institutes of Health, bethesda, MD
2-Al-Lazikani et Al, (1997) JMB 273,927-948
Thus, unless otherwise specified, it is to be understood that a "CDR" or "complementarity determining region" or a separately specified CDR (e.g., CDR-H1, CDR-H2) of a given antibody or region thereof (e.g., variable region thereof) encompasses one (or a particular) complementarity determining region as defined by any of the above schemes. For example, in stating that a particular CDR (e.g., CDR-H3) contains a given V H Or V L In the case of the amino acid sequence of a corresponding CDR in the amino acid sequence, it is to be understood that such CDR has the sequence of the corresponding CDR (e.g., CDR-H3) within the variable region, as defined by any of the above schemes. In some embodiments, specified CDR sequences are specified.
Likewise, unless specified otherwise, it is to be understood that the FR of a given antibody or region thereof (e.g., variable region thereof) or a separately specified FR(s) (e.g., FR-H1, FR-H2) encompasses one (or a specific) framework region as defined by any known scheme. In some cases, schemes are specified for identifying a particular CDR, FR, or FR or CDR, such as the CDR defined by Kabat, chothia or Contact method. In other cases, specific amino acid sequences of CDRs or FR are given.
The term "variable region" or "variable domain" refers to a domain of an antibody that is involved in the binding of the antibody to an antigen in the heavy or light chain of the antibody. The variable domains of the heavy and light chains of natural antibodies (V respectively H And V L ) Typically having a similar structure, each domain comprises four conserved Framework Regions (FR) and three CDRs. (see, e.g., kit et al Kuby Immunology, 6 th edition, w.h. freeman and co., p 91 (2007). Singular V) H Or V L The domain may be sufficient to confer antigen binding specificity. In addition, V from antigen-binding antibodies can be used H Or V L Domain isolation of antibodies binding to specific antigens for separate selection of complementary V L Or V H Library of domains. See, e.g., portolano et al, J.Immunol.150:880-887 (1993); clarkson et al Nature 352:624-628 (1991).
Antibodies provided include antibody fragments. An "antibody fragment" refers to a molecule other than an intact antibody that comprises a portion of the intact antibody that binds to an antigen to which the intact antibody binds. Examples of antibody fragments include, but are not limited to Fv, fab, fab ', fab ' -SH, F (ab ') 2 The method comprises the steps of carrying out a first treatment on the surface of the A diabody; a linear antibody; single chain antibody molecules (e.g., scFv); and multispecific antibodies formed from antibody fragments. In particular embodiments, the antibody is a single chain antibody fragment, such as an scFv, comprising a variable heavy chain region and/or a variable light chain region.
A single domain antibody is an antibody fragment comprising all or part of the heavy chain variable domain or all or part of the light chain variable domain of an antibody. In certain embodiments, the single domain antibody is a human single domain antibody.
Antibody fragments may be prepared by a variety of techniques including, but not limited to, proteolytic digestion of intact antibodies and production by recombinant host cells. In some embodiments, the antibodies are recombinantly produced fragments, such as fragments comprising a naturally non-occurring arrangement (e.g., those having two or more antibody regions or chains joined by a synthetic linker (e.g., a peptide linker), and/or fragments that are not produced by enzymatic digestion of a naturally occurring intact antibody. In some aspects, the antibody fragment is an scFv.
A "humanized" antibody is one in which all or substantially all CDR amino acid residues are derived from non-human CDRs and all or substantially all Framework Region (FR) amino acid residues are derived from human FRs. In some embodiments, the humanized form of a non-human antibody (e.g., a murine antibody) is a chimeric antibody that contains minimal sequences derived from a non-human immunoglobulin. In certain embodiments, the humanized antibody is an antibody from a non-human species that has one or more Complementarity Determining Regions (CDRs) from the non-human species and a Framework Region (FR) from a human immunoglobulin molecule. In some embodiments, the humanized antibody optionally may comprise at least a portion of an antibody constant region derived from a human antibody. "humanized form" of a non-human antibody refers to a variant of a non-human antibody that has undergone humanization to generally reduce immunogenicity to humans, while retaining the specificity and affinity of the parent non-human antibody. In some embodiments, some FR residues in a humanized antibody are substituted with corresponding residues from a non-human antibody (e.g., an antibody from which CDR residues are derived), e.g., to restore or improve antibody specificity or affinity. (see, e.g., U.S. Pat. No. 5,585,089 to Queen and U.S. Pat. No. 5,225,539 to Winter). Such chimeric monoclonal antibodies and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art.
In certain embodiments, the humanized antibody is a human immunoglobulin (recipient antibody) in which residues from the heavy chain variable region of the recipient are replaced with residues from the heavy chain variable region of a non-human species (donor antibody), such as a mouse, rat, rabbit, or non-human primate having the desired specificity, affinity, and/or capacity. In some cases, FR residues of the human immunoglobulin are replaced with corresponding non-human residues. In addition, the humanized antibody may comprise residues not found in the recipient antibody or the donor antibody. In some embodiments, the nucleic acid sequences encoding the human variable heavy and variable light chains are altered to replace one or more CDR sequences of the human (acceptor) sequence with sequences encoding corresponding CDRs in the non-human antibody sequence (donor sequence). In some embodiments, the human receptor sequence may comprise FR derived from a different gene. In a particular embodiment, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR are those of a human immunoglobulin sequence. In some embodiments, the humanized antibody will optionally also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details, see, e.g., jones et al, nature 321:522-525 (1986); riechmann et al Nature 332:323-329 (1988); and Presta, curr.Op.struct.biol.2:593-596 (1992). See also, for example, vaswani and Hamilton, ann. Allergy, asthma & Immunol.1:105-115 (1998); harris, biochem. Soc. Transactions 23:1035-1038 (1995); hurle and Gross, curr.op.Biotech.5:428-433 (1994); and U.S. patent nos. 6,982,321 and 7,087,409, which are incorporated herein by reference. In some embodiments, provided herein are humanized anti-idiotype antibodies.
In certain embodiments, the antibody (e.g., an anti-idiotype antibody) is humanized. In certain embodiments, the antibodies are humanized by any suitable known method. For example, in some embodiments, a humanized antibody may have incorporated therein one or more amino acid residues of non-human origin. These non-human amino acid residues are often referred to as "import" residues, which are typically taken from an "import" variable domain. In particular embodiments, humanization may be achieved substantially by following the method of Winter and colleagues (Jones et al (1986) Nature 321:522-525; riechmann et al (1988) Nature 332:323-327; verhoeyen et al (1988) Science 239:1534-1536), such as by substituting hypervariable region sequences for the corresponding sequences of human antibodies. Thus, such "humanized" antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567) in which significantly less than the complete human variable domain is replaced with the corresponding sequence of a non-human species. In certain embodiments, the humanized antibody is a human antibody in which some hypervariable region residues and possibly some FR residues are substituted with residues from similar sites in a rodent antibody.
Sequences encoding full length antibodies can then be obtained by ligating the provided variable heavy and variable light chain sequences to human constant heavy and constant light chain regions. Suitable human constant light chain sequences include kappa and lambda constant light chain sequences. Suitable human constant heavy chain sequences include IgG1, igG2, and sequences encoding IgG1 mutants having the presented immunostimulatory properties. Such mutants may have reduced ability to activate complement and/or antibody dependent cytotoxicity and are described in U.S. Pat. No. 5,624,821, WO 99/58372, U.S. Pat. No. 6,737,056. Suitable constant heavy chains also include IgG1 comprising a deletion of substitution E233P, L234V, L235A, A327G, A330S, P331S and residue 236. In another embodiment, the full length antibody comprises IgA, igD, igE, igM, igY or IgW sequences.
Suitable human donor sequences can be determined by the following method: the peptide sequence encoded by the mouse donor sequence is compared to a peptide sequence encoded by a set of human sequences, preferably sequences encoded by human germline immunoglobulin genes or mature antibody genes. Human sequences with high sequence homology, preferably with the highest homology determined, can serve as acceptor sequences for the humanization process.
In addition to exchanging human CDRs for mouse CDRs, further manipulations can be performed in human donor sequences to obtain sequences encoding humanized antibodies with optimized properties (e.g., affinity for antigen).
In addition, the altered human acceptor antibody variable domain sequences may also be provided to encode one or more amino acids corresponding to positions 4, 35, 38, 43, 44, 46, 58, 62, 64, 65, 66, 67, 68, 69, 73, 85, 98 of the light chain variable region and positions 2, 4, 36, 39, 43, 45, 69, 70, 74, 75, 76, 78, 92 of the heavy chain variable region of the non-human donor sequence (according to the Kabat numbering system) (Carter and Presta, U.S. patent No. 6,407,213).
In particular embodiments, it is generally desirable that the antibodies retain high affinity for the antigen and other favorable biological properties after humanization. In some embodiments, to achieve this, humanized antibodies are prepared by a process of analyzing a parent sequence and various conceptual humanized products using a three-dimensional model of the parent sequence and humanized sequence. Three-dimensional models of immunoglobulins are generally available and familiar to those skilled in the art. A computer program is available that illustrates and displays the possible three-dimensional conformational structure of the selected candidate immunoglobulin sequence. By examining these displays allows analysis of the possible role of the residues in the functioning of the candidate immunoglobulin sequence, i.e. analysis of residues affecting the ability of the candidate immunoglobulin to bind its antigen. In this way, FR residues can be selected from the receptor sequence and the input sequence and combined to obtain the desired antibody characteristics, such as increased affinity for one or more target antigens. Typically, the hypervariable region residues are directly and most essentially involved in the effect of antigen binding.
In certain embodiments, the selection of both human light and heavy chain variable domains for use in preparing humanized antibodies may be important for reducing antigenicity. The entire library of known human variable domain sequences is screened with the variable domain sequences of rodent antibodies according to the so-called "best fit" method. The human sequence closest to the rodent is then accepted as the human framework for the humanized antibody. See, for example, sims et al (1993) J.Immunol.151:2296; chothia et al (1987) J.mol.biol.196:901. Another approach uses a specific framework of consensus sequences of all human antibodies derived from a specific light chain or heavy chain subgroup. The same framework can be used for several different humanized antibodies. See, e.g., carter et al (1992) Proc. Natl. Acad. Sci. USA,89:4285; presta et al (1993) J.Immunol.,151:2623.
Antibodies provided include human antibodies. A "human antibody" is an antibody having an amino acid sequence that corresponds to the amino acid sequence of a human or human cell or an antibody produced from a non-human source using a human antibody repertoire or other human antibody coding sequence (including a human antibody library). The term excludes humanized versions of non-human antibodies that comprise non-human antigen binding regions, such as those in which all or substantially all CDRs are non-human.
Human antibodies can be prepared by administering an immunogen to a transgenic animal that has been modified to produce a fully human antibody or a fully antibody with human variable regions in response to antigen challenge. Such animals typically contain all or part of the human immunoglobulin locus, either replacing the endogenous immunoglobulin locus or which is present extrachromosomally or randomly integrated into the chromosome of the animal. In such transgenic animals, typically the endogenous immunoglobulin loci have been inactivated. Human antibodies may also be derived from human antibody libraries containing antibody coding sequences derived from human libraries, including phage display and cell-free libraries.
Antibodies provided include monoclonal antibodies, including monoclonal antibody fragments. The term "monoclonal antibody" as used herein refers to an antibody obtained from or within a population of substantially homogeneous antibodies (i.e., the individual antibodies comprising the population are identical except for possible variants that contain naturally occurring mutations or that are produced during production of a monoclonal antibody preparation, such variants typically being present in minor amounts). In contrast to polyclonal antibody preparations, which typically include different antibodies directed against different epitopes, each monoclonal antibody of a monoclonal antibody preparation is directed against a single epitope on an antigen. The term should not be construed as requiring the production of antibodies by any particular method. Monoclonal antibodies can be prepared by a variety of techniques including, but not limited to, from hybridoma production, recombinant DNA methods, phage display, and other antibody display methods.
2. Target expressing cells
In some embodiments, the recombinant receptor stimulant is a cell that expresses a target recognized by an antigen receptor, i.e., the recombinant receptor stimulant is a target expressing cell. In some embodiments, the target is an antigen of a recombinant receptor, and thus in some cases, the target-expressing cell is an antigen-expressing cell. In some embodiments, the recombinant receptor stimulant is an antigen-expressing cell, such as a cell that expresses an antigen as described above.
In certain embodiments, the cell (e.g., a target expressing cell, such as an antigen expressing cell) is exogenous, heterologous, and/or autologous to the subject. In some embodiments, the cell is exogenous to the subject.
In certain embodiments, the target expressing cells express a target bound and/or recognized by a recombinant receptor. In some embodiments, the target is an antibody and the target expressing cell expresses the antibody. In some embodiments, the target expressing cell is a tumor cell. In certain embodiments, the target expressing cell is a primary cell.
In some embodiments, the target is an antigen recognized by a recombinant receptor, and the target-expressing cell is an antigen-expressing cell. In certain embodiments, the antigen-expressing cells express an antigen that is bound and/or recognized by a recombinant receptor. In some embodiments, the antigen expressing cell is a tumor cell. In certain embodiments, the antigen expressing cell is a primary cell. In some embodiments, the cell line is an immortal cell line. In certain embodiments, the antigen-expressing cells are cancerous and/or tumor cells. In some embodiments, the antigen-expressing cells are derived from cancer cells and/or tumor cells, e.g., human cancer cells and/or human tumor cells. In some embodiments, the antigen expressing cell is a cell from a cancer cell line, optionally a human cancer cell line. In some embodiments, the antigen expressing cells are cells from a tumor cell line, optionally a human tumor cell line.
In a particular embodiment, the antigen expressing cell is a tumor cell. In some embodiments, the antigen-expressing cell is a circulating tumor cell, e.g., a neoplastic immune cell, such as a neoplastic B cell (or a cell derived from a neoplastic B cell).
In particular embodiments, the antigen expressing cells express integrin (avb 6 integrin), B Cell Maturation Antigen (BCMA), B7-H3, B7-H6, carbonic anhydrase 9 (CA 9, also known as CAIX or G250), cancer-testis antigen, cancer/testis antigen IB (CTAG, also known as NY-ESO-1 and rage-2), carcinoembryonic antigen (CEA), cyclin A2, C-C motif chemokine ligand 1 (CCL-1), CD19, CD20, CD22, CD23, CD24, CD30, CD33, CD38, CD44v6, CD44v7/8, CD123, CD133, CD138, CD171, chondroitin sulfate proteoglycan 4 (CSPG 4), epidermal growth factor protein (EGFR), truncated epidermal growth factor protein (tgfr), growth factor receptor type III mutant epidermal glycoprotein 2 (EPG-2), epidermal glycoprotein 40 (EPG-40), liver ligand B2, hepadulter 2, fcreceptor 5 (Fc-5), and the like; also known as Fc receptor homolog 5 or FCRH 5), fetal acetylcholine receptor (fetal AchR), folic acid binding protein (FBP), folic acid receptor alpha, fetal acetylcholine receptor, ganglioside GD2, O-acetylated GD2 (OGD 2), ganglioside GD3, glycoprotein 100 (gp 100), glypican-3 (GPC 3), G-protein coupled receptor 5D (GPCR 5D), her2/neu (receptor tyrosine kinase erb-B2), her3 (erb-B3), her4 (erb-B4), erbB dimer, human high molecular weight melanomA-Associated antigen (HMW-MAA), hepatitis B surface antigen, human leukocyte antigen A1 (HLA-AIA 1), human leukocyte antigen A2 (HLa-A2), IL-22 receptor alpha (IL-22 Ra), IL-13 receptor alpha 2 (IL-13 Ra 2), kinase insert domain receptor (kdr), kappa light chain, L1 cell adhesion molecule (L1-CAM), CE7 epitope of L1-CAM, leucine rich repeat containing protein 8 family member a (LRRC 8A), lewis Y, melanomA-Associated antigen (MAGE) -Al MAGE-A3, MAGE-A6, MAGE-a10, mesothelin (MSLN), c-Met, murine Cytomegalovirus (CMV), mucin 1 (MUC 1), MUC16, natural killer cell group 2 member D (KG 2D) ligands, melanin a (MART-1), neural Cell Adhesion Molecules (NCAM), carcinoembryonic antigen, melanoma preferential expression antigen (PRAME), progesterone receptor, prostate specific antigen, prostate Stem Cell Antigen (PSCA), prostate Specific Membrane Antigen (PSMA), receptor tyrosine kinase-like orphan receptor 1 (ROR 1), survivin, trophoblast glycoprotein (TPBG, also known as 5T 4), tumor associated glycoprotein 72 (TAG 72), tyrosinase associated protein 1 (TRP 1, also known as TYRP1 or gp 75), tyrosinase related protein 2 (TRP 2, also known as dopachrome tautomerase, dopachrome delta isomerase, or DCT), vascular Endothelial Growth Factor Receptor (VEGFR), vascular endothelial growth factor receptor 2 (VEGFR 2), wilms tumor 1 (WT-1), or a combination thereof. In some embodiments, the antigen expressing cells express antigens specific for or expressed by the pathogen, or antigens associated with a universal tag, and/or biotinylated molecules, and/or molecules expressed by HIV, HCV, HBV or other pathogens. In certain embodiments, the antigen-expressing cells express one or more antigens associated with a B cell malignancy, such as any of a number of known B cell markers. In certain embodiments, the antigen expressing cells express CD20, CD19, CD22, ROR1, CD45, CD21, CD5, CD33, igκ, igλ, CD79a, CD79b, CD30, or a combination thereof. In some embodiments, the antigen expressing cells express CD19, e.g., human CD19.
In some embodiments, the antigen is or includes an antigen that is characteristic of or expressed by a pathogen. In some embodiments, the antigen is a viral antigen (e.g., a viral antigen from HIV, HCV, HBV, etc.), a bacterial antigen, and/or a parasitic antigen. In certain embodiments, the antigen expressing cell is or is derived from a tumor cell. In some embodiments, the tumor cell is cancerous. In certain embodiments, the tumor cells are non-cancerous. In some embodiments, the tumor cells are or are derived from circulating B cells, such as circulating B cells that are capable of forming tumors in vivo. In some embodiments, the tumor cell is or is derived from a circulating B cell that is a neoplastic, tumorigenic, or cancerous B cell.
In certain embodiments, the tumor cell is or is derived from a human cancer cell. In some embodiments, the tumor cells are derived from cells of the following cancers: AIDS-related cancers, breast cancer, gut/gastrointestinal cancer, anal cancer, appendiceal cancer, cholangiocarcinoma, colon cancer, colorectal cancer, esophageal cancer, gallbladder cancer, islet cell tumor, pancreatic neuroendocrine tumor, liver cancer, pancreatic cancer, rectal cancer, small intestine cancer, stomach (gastric) cancer, cancer of the endocrine system, adrenocortical cancer, parathyroid cancer, pheochromocytoma, pituitary tumor, thyroid cancer, eye cancer, intraocular melanoma, retinoblastoma, bladder cancer, renal (renal cell) cancer, penile cancer, prostate cancer, transitional cell renal pelvis and ureter cancer, testicular cancer, urethral cancer, nephroblastoma or other childhood renal tumors, blastoma, central nervous system cancer, extracranial blastoma, extragonadal blastoma, ovarian blastoma, gynaecological cancer, cervical cancer, endometrial cancer gestational trophoblastic tumors, ovarian epithelial cancers, uterine sarcomas, vaginal cancers, vulvar cancers, head and neck cancers, hypopharyngeal cancers, laryngeal cancers, lip and oral cancers, metastatic squamous neck cancers (metastatic squamous neck cancer), nasopharyngeal cancers, oropharyngeal cancers, sinus and nasal cancers, pharyngeal cancers, salivary gland cancers, pharyngeal cancers, musculoskeletal cancers, bone cancers, ewing's sarcoma, gastrointestinal stromal tumors (GIST), osteosarcomas, osteomalignant fibrous histiocytomas, rhabdomyosarcomas, soft tissue sarcomas, uterine sarcomas, nervous system cancers, brain tumors, astrocytomas, brain stem gliomas, central nervous system atypical teratomas/rhabdomyomas, central nervous system embryonomas, craniopharyngemas, ependymomas, neuroblastomas, spinal cord tumors, supracurtain primitive neuroectodermal tumors and pineal blastomas, neuroblastoma, respiratory cancer, thymus cancer, non-small cell lung cancer, malignant mesothelioma, thymoma, thymus cancer, skin cancer, kaposi's sarcoma, melanoma, or merkel cell cancer, or any equivalent human cancer thereof.
In certain embodiments, the tumor cells are derived from a non-hematologic cancer, e.g., a solid tumor. In certain embodiments, the tumor cells are derived from hematologic cancers. In certain embodiments, the tumor cells are derived from cancer that is a B cell malignancy or hematological malignancy. In particular embodiments, the tumor cells are derived from non-hodgkin's lymphoma (NHL), acute Lymphoblastic Leukemia (ALL), chronic Lymphoblastic Leukemia (CLL), diffuse large B-cell lymphoma (DLBCL), acute Myelogenous Leukemia (AML), or myeloma (e.g., multiple Myeloma (MM)), or any equivalent human cancer thereof. In some embodiments, the antigen expressing cells are neoplastic, cancerous, and/or tumorigenic B cells. A variety of tumor cell lines are known and available and can be selected based on the antigen recognized by a particular recombinant receptor (e.g., CAR).
Any of a number of tumour cell lines are known and available. Tumor cell lines expressing a particular tumor antigen are known or surface expression of the tumor antigen can be readily determined or measured by the skilled artisan using any of a variety of techniques, such as by flow cytometry. Exemplary tumor cell lines include, but are not limited to, lymphoma cells (Raji; daudi; jeko-1; BJAB; ramos; NCI-H929; BCBL-1; DOHH-2, SC-1, WSU-NHL, JVM-2, rec-1, SP-53, RL, granta 519, NCEP-1, CL-01); leukemia cells (BALL-1, RCH-ACV, SUP-B15); cervical cancer cells (33A; caSki; heLa); lung cancer cells (NCI-H358; A549, H1355, H1975, calu-1, H1650 and H727); breast cancer cells (Hs-578T; ZR-75-1; MCF-7/HER2; MCF10A; MDA-MB-231; SKBR-3, BT-474, MDA-MB-231); ovarian cancer cells (ES-2; SKOV-3; OVCAR3; HEY 1B); multiple myeloma cells (U266, NCI-H929, RPMI-8226, OPM2, LP-1, L363, MM.1S, MM.1R, MC/CAR, JJN3, KMS11, AMO-1, EJM; MOLP-8). For example, exemplary CD19 expressing cell lines include, but are not limited to Raji, daudi, and BJAB; exemplary CD20 expressing cell lines include Daudi, ramos, and Raji; exemplary CD22 expressing cell lines include, but are not limited to Ramos, raji, A549, H727, and H1650; exemplary Her2 expressing cell lines include SKOV3, BT-474 and SKBR-3; exemplary BCMA expressing cell lines include, but are not limited to, RPMI-8226, NCI-H929, MM1S, MM R, and KMS11; exemplary GPRC 5D-expressing cell lines include, but are not limited to, AMO-1, EJM, NCI-H929, MM.1S, MM1.R, MOLP-8, and OPM-2; exemplary ROR1 expressing cell lines include, but are not limited to, A549, MDA-MB-231, H1975, BALL-1, and RCH-ACV.
In some embodiments, the target expressing cell line is a cell line that has been transduced to express a target of a recombinant receptor. In some embodiments, the target is a tumor antigen. In a particular embodiment, the antigen expressing cell line is a cell line that has been transduced to express a tumor antigen. The cell line may be a mammalian cell line including, but not limited to, a human cell line. In some embodiments, the human cell line may be K562, U937, 721.221, T2, and C1R cells. For example, nucleic acids encoding tumor antigens may be introduced into the K562 chronic myelogenous leukemia cell line. In some embodiments, the cell line may be engineered with a plasmid vector or messenger RNA (mRNA) encoding the tumor antigen of interest. In some embodiments, the introducing may be by lentivirus-based transduction. In some embodiments, the cell line (e.g., K562 cells) stably expresses the exogenous nucleic acid encoding the tumor antigen. In some embodiments, the exogenous nucleic acid may be integrated into the genome of a cell line (e.g., a K562 cell). In some embodiments, the exogenous nucleic acid may be integrated at a particular locus in the genome of a cell line (e.g., a K562 cell). In some embodiments, the exogenous nucleic acid may be integrated into the genome of a cell line (e.g., K562 cells) at genomic safe harbor (genomic safe harbour, GSH). GSH is a site that supports stable integration and expression of exogenous nucleic acids while minimizing the risk of undesired interactions with the host cell genome (see, e.g., sadelain et al, nat Rev cancer (2011) 12 (1): 51-8). Several safe GSH for stable integration of exogenous nucleic acids into human cells have been identified, including AAVS1, a naturally occurring integration site of AAV virus on chromosome 19; CCR5 gene, a chemokine receptor gene, also known as HIV-1 co-receptor; and human ortholog of the mouse Rosa26 locus (see, e.g., papapetrou and Schambach Mol ter (2016) 24 (4): 678-684).
In some embodiments, the target-expressing cells are varied or stepwise adjusted at different rates over multiple incubations as compared to a fixed amount of cells (effector cells) expressing the therapeutic composition of the recombinant receptor. In some embodiments, the step-wise adjustment amount is from 100:1 to 0.001 ratio of target-expressing target cells to effector T cells (T: E), such as from 50:1 to 0.050 ratio of T: E, from 25:1 to 0.025 ratio of T: E, from 12:1 to 0.012:1 ratio of T: E, from 10:1 to 0.010 ratio of T: E, or from 5:1 to 0.5 ratio of T: E. In some embodiments, the ratio is or is a ratio of T to E of about from 12:1 to 0.012:1. The specific ratio ranges may be empirically determined based on the specific target and target cells employed. For example, the selected ratio is a ratio that includes a linear dose response increase in recombinant receptor-dependent activity across a plurality of stepwise adjustments. In some embodiments, the ratio is selected to further include a lower asymptote for receptor-dependent activity and a higher asymptote for receptor-dependent activity, which represent minimum and maximum responses, respectively.
For example, the target is an antigen of a recombinant receptor. In some embodiments, antigen expressing cells are varied or stepwise adjusted at different rates over multiple incubations as compared to a fixed amount of cells (effector cells) expressing a therapeutic composition of the recombinant receptor. In some embodiments, the stepwise adjustment amount is from 100:1 to 0.001 ratio of antigen expressing target cells to effector T cells (T: E), such as from 50:1 to 0.050 ratio of T: E, from 25:1 to 0.025 ratio of T: E, from 12:1 to 0.012:1 ratio of T: E, from 10:1 to 0.010 ratio of T: E, or from 5:1 to 0.5 ratio of T: E. In some embodiments, the ratio is or is a ratio of T to E of about from 12:1 to 0.012:1. The specific ratio ranges may be empirically determined based on the specific antigen and target cell employed. For example, the selected ratio is a ratio that includes a linear dose response increase in recombinant receptor-dependent activity across a plurality of stepwise adjustments. In some embodiments, the ratio is selected to further include a lower asymptote for receptor-dependent activity and a higher asymptote for receptor-dependent activity, which represent minimum and maximum responses, respectively.
C. Measurement of recombinant receptor dependent Activity
The methods provided herein for assessing efficacy include measuring the activity of a therapeutic cell composition in response to stimulation of a recombinant receptor of an engineered cell of the therapeutic cell composition. As described above, the provided assays allow measurement of recombinant receptor-dependent activity in response to recombinant receptor stimulators (as described in section I-B) from a plurality of incubation conditions, wherein each incubation includes cells of a different stepwise adjustment ratio of therapeutic cell composition to recombinant receptor stimulators.
In particular embodiments, it is contemplated that the recombinant receptor-dependent activity (e.g., CAR-dependent activity) is an activity that occurs in an engineered cell that expresses the recombinant receptor, that is not and/or cannot occur in a cell that does not express the recombinant receptor. In some embodiments, the recombinant receptor-dependent activity is an activity that depends on or is present at the recombinant receptor. Recombinant receptor-dependent activity may be any cellular process that is directly or indirectly affected by expression and/or presence of a recombinant receptor or by a change in activity of a recombinant receptor (e.g., receptor stimulation). In some embodiments, the recombinant receptor-dependent activity may include, but is not limited to, cellular processes such as cell division, DNA replication, transcription, protein synthesis, membrane transport, protein translocation and/or secretion, or it may be an immune cell function, such as cytolytic activity. In certain embodiments, recombinant receptor-dependent activity can be measured by confirmation of CAR receptors, phosphorylation of intracellular signaling molecules, degradation of proteins, transcription, translation, translocation of proteins, and/or changes in production and secretion of factors (e.g., proteins, or growth factors, cytokines). In certain embodiments, the recombinant receptor is a CAR. In certain embodiments, the recombinant receptor is a TCR.
In some embodiments, the recombinant receptor-dependent activity (e.g., CAR-dependent activity) is a measure of a factor, such as an amount or concentration, or a change in an amount or concentration, after stimulation of the therapeutic cell composition with the recombinant receptor stimulator. In certain embodiments, the factor may be a protein, a phosphorylated protein, a cleaved protein, an translocated protein, a protein in activity confirmation, a polynucleotide, an RNA polynucleotide, an mRNA, and/or an shRNA. In some embodiments, the measurement may include, but is not limited to, an increase or decrease in kinase activity, protease activity, phosphatase activity, cAMP production, ATP metabolism, translocation (e.g., nuclear localization of a protein), an increase in transcriptional activity, an increase in translational activity, production and/or secretion of soluble factors, cellular uptake, ubiquitination, and/or protein degradation.
In some embodiments, the factor is a secreted soluble factor, such as a hormone, a growth factor, a chemokine, and/or a cytokine.
In some embodiments, the recombinant receptor-dependent activity (e.g., CAR-dependent activity) is a response to stimulation with a recombinant receptor stimulator. In certain embodiments, the cells are incubated in the presence of a recombinant receptor stimulator capable of stimulating recombinant receptor-dependent activity, and the activity is or includes at least one aspect of a response to the stimulus. The response may include, but is not limited to, an intracellular signaling event (e.g., an increase in receptor molecule activity, an increase in kinase activity of one or more kinases, an increase in transcription of one or more genes, an increase in protein synthesis of one or more proteins), and/or an intracellular signaling molecule (e.g., an increase in kinase activity of a protein). In some embodiments, the response (e.g., recombinant receptor-dependent activity) is associated with immune activity, and may include, but is not limited to, soluble factor (e.g., cytokine) production and/or secretion, increased antibody production, and/or increased cytolytic activity.
In some embodiments, recombinant receptor-dependent activity is assessed by measuring, detecting, or quantifying recombinant receptor-dependent activity (i.e., at least one activity that is initiated, triggered, supported, prolonged, and/or caused by a stimulus (e.g., a recombinant receptor stimulator)) against a stimulus (e.g., a recombinant receptor stimulator). In certain embodiments, the cells of the therapeutic cell composition are incubated with a recombinant receptor stimulator, wherein the interaction or binding of the recombinant receptor stimulator with the recombinant receptor stimulates (e.g., induces) recombinant receptor-dependent activity specific for cells expressing the recombinant receptor. In certain embodiments, the recombinant receptor-dependent activity occurs in cells that express the recombinant receptor, but not in cells that do not express the receptor or only minimally in cells that do not express the receptor. In a particular embodiment, the recombinant receptor is a CAR. In some embodiments, the activity is CAR-dependent activity.
Conditions for stimulating a recombinant receptor of an engineered cell (e.g., immune cell or T cell) by a recombinant receptor stimulator may include one or more of the following: specific media, temperature, oxygen content, carbon dioxide content, time, agents (e.g., nutrients, amino acids, antibiotics, ions). In some embodiments, recombinant receptor-dependent activity is determined by whether a soluble factor (e.g., a cytokine or chemokine) is produced or secreted.
In some embodiments, the recombinant receptor-dependent activity is specific for a cell expressing the recombinant receptor. In some embodiments, the recombinant receptor-dependent activity is specific for a cell expressing the recombinant receptor and does not occur in a cell lacking expression of the recombinant receptor. In certain embodiments, the recombinant receptor is a CAR and the activity is CAR-dependent activity. In certain embodiments, the activity is not present in a cell lacking expression of the recombinant receptor under the same conditions that recombinant receptor-dependent activity is present in a cell expressing the recombinant receptor. In certain embodiments, the CAR-dependent activity is about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 97%, about 98%, about 99%, or about 99% less than the CAR-dependent activity of the CAR cell under the same conditions.
In some embodiments, the recombinant receptor-dependent activity is specific for a cell expressing a recombinant receptor (e.g., CAR), and the activity is generated by stimulation with a recombinant receptor stimulator specific for cells of a therapeutic cellular composition expressing the recombinant receptor. In some embodiments, the recombinant receptor is a CAR, and the CAR specifically stimulates, triggers, initiates, induces and/or prolongs the activity of the car+ cells, but does not stimulate, trigger, initiate, induce and/or prolong the activity of the CAR-cells. In some embodiments, the CAR-dependent activity is about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95%, about 97%, about 98%, about 99%, or about 99% less in the CAR-cells than in the car+ cells after stimulation by the CAR-specific stimulus.
In certain embodiments, the activity is a recombinant receptor-dependent (e.g., CAR-dependent) activity stimulated by a recombinant receptor stimulator specific for the recombinant receptor (as described in section I-B). In some embodiments, the recombinant receptor stimulator (e.g., CAR-specific agent) comprises an antigen or epitope thereof that is bound and/or recognized by a recombinant receptor (e.g., CAR). In some embodiments, the recombinant receptor stimulant includes an antibody that binds to a recombinant receptor (e.g., an anti-idiotype antibody (anti-ID)) or an active fragment, variant, or portion thereof. In certain embodiments, the recombinant receptor stimulant is a cell that expresses an antigen on its surface. In certain embodiments, the recombinant receptor stimulant is a cell that expresses an antibody on its surface. In some embodiments, the cells are from a cell line, as described in section I-B-2. In some embodiments, the cell line is a tumor cell line. In some embodiments, the cell expresses a tumor antigen.
In some embodiments, recombinant receptor-dependent activity is measured in a therapeutic cell composition containing cells expressing a recombinant receptor (e.g., CAR), and the measurement is compared to one or more controls. In certain embodiments, the control is a similar or identical cell composition that is not stimulated. For example, in some embodiments, recombinant receptor-dependent activity is measured in a cell composition after or during incubation with a recombinant receptor stimulator, and the resulting measurement is compared to a control measurement of activity from a similar or identical cell composition not incubated with the recombinant receptor stimulator. In some embodiments, both the therapeutic cell composition and the control cell composition contain cells that express the recombinant receptor. In some embodiments, the control is taken from a similar cell composition that does not contain cells expressing the recombinant receptor (e.g., car+ cells). Thus, in some embodiments, a therapeutic cell composition containing recombinant receptor expressing cells and a control cell composition not containing recombinant receptor expressing cells are contacted with a recombinant receptor stimulator. In certain embodiments, the control is a measurement from the same cellular composition expressing the recombinant receptor taken prior to any stimulation. In certain embodiments, a control measurement is obtained to determine the background signal, and the control measurement is subtracted from the measure of activity. In some embodiments, the measured value of activity in the cell composition is divided by the control measured value to obtain a value of the ratio of activity to control level. In some embodiments, all recombinant receptor-dependent activity measurements are adjusted or normalized relative to a control measurement (e.g., wherein the recombinant receptor stimulant is not cultured with cells of the therapeutic cell composition). In some embodiments, adjusting or normalizing the measurement relative to control conditions provides a more accurate measure of recombinant receptor-dependent activity.
In particular embodiments, the recombinant receptor-dependent activity is or includes the production and/or secretion of soluble factors. In some embodiments, the recombinant receptor (e.g., CAR) -dependent activity is or includes the production and/or secretion of soluble factors. In certain embodiments, the soluble factor is a cytokine or chemokine.
Suitable techniques for measuring the production or secretion of soluble factors are known in the art. The production and/or secretion of a soluble factor may be measured by determining the concentration or amount of the extracellular amount of the factor, or determining the amount of transcriptional activity of a gene encoding the factor. Suitable techniques include, but are not limited to, the following assays: such as immunoassays, aptamer-based assays, histological or cytological assays, mRNA expression level assays, enzyme-linked immunosorbent assays (ELISA), immunoblots, immunoprecipitations, radioimmunoassays (RIA), immunostaining, flow cytometry assays, surface Plasmon Resonance (SPR), chemiluminescent assays, lateral flow immunoassays, inhibition assays or affinity assays, protein microarrays, high Performance Liquid Chromatography (HPLC), meso Scale Discovery (MSD) electrochemiluminescence, and bead-based Multiplex Immunoassays (MIA). In some embodiments, suitable techniques may use a detectable binding reagent that specifically binds to the soluble factor.
In certain embodiments, the measurement of the soluble factor (e.g., cytokine) is measured by ELISA (enzyme linked immunosorbent assay). ELISA is a plate-based assay technology designed to detect and quantify substances such as peptides, cytokines, antibodies, and hormones. In ELISA, soluble factors must be immobilized on a solid surface and then complexed with an antibody linked to an enzyme. Detection is accomplished by assessing conjugated enzyme activity via incubation with a substrate to produce a detectable signal. In some embodiments, the recombinant receptor-dependent activity is measured using an ELISA assay.
In some embodiments, the recombinant receptor-dependent activity is secretion or production of a soluble factor (e.g., a cytokine). In certain embodiments, production or secretion is stimulated in a therapeutic cell composition containing recombinant receptor-expressing cells (e.g., CAR-expressing cells) by a recombinant receptor stimulator capable of binding to a recombinant receptor to stimulate recombinant receptor-dependent activity (e.g., CAR-dependent activity). In some embodiments, the recombinant receptor stimulant comprises an antigen or epitope thereof that is specific for a recombinant receptor; is a cell expressing an antigen; or an antibody or a portion or variant thereof that binds to and/or recognizes a recombinant receptor; or a combination thereof (see, e.g., section I-B above). In certain embodiments, the recombinant receptor stimulant is a recombinant protein that comprises an antigen, or epitope thereof, bound or recognized by a recombinant receptor.
In certain embodiments, the recombinant receptor-dependent activity is soluble factor production and/or secretion as measured by incubating a therapeutic cell composition containing cells expressing a recombinant receptor (e.g., CAR) with a recombinant receptor stimulator (as described in section I-B). In certain embodiments, the soluble factor is a cytokine or chemokine. In some embodiments, cells containing recombinant receptor expressing cells in a therapeutic cell composition are incubated in the presence of a recombinant receptor stimulator for an amount of time, and the production and/or secretion of soluble factors is measured at one or more time points during the incubation. In some embodiments, the cells are incubated with the recombinant receptor stimulant for a duration of up to or about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 18 hours, about 19 hours, about 20 hours, about 21 hours, about 22 hours, about 23 hours, about 24 hours, about 48 hours, or between 1 hour and 4 hours, between 1 hour and 12 hours, between 12 hours and 24 hours (each inclusive), or more than 24 hours, and the amount of soluble factor (e.g., cytokine) is detected.
In some embodiments, the recombinant receptor stimulant is a particle (e.g., a bead) to which an antigen or portion thereof recognized by the recombinant receptor is attached or immobilized, or to which an antibody (e.g., an anti-idiotype antibody) specific for an extracellular domain of the recombinant receptor (e.g., an extracellular antigen binding domain (e.g., scFv)) is attached or immobilized. In some embodiments, a constant number of cells of the recombinant receptor (e.g., CAR) and therapeutic cell composition are incubated with the particles at a plurality of ratios of cells of the therapeutic cell composition to particles, such as including ratios of or about 1:100, 1:75, 1:50, 1:40, 1:30, 1:20, 1:15, 1:14, 1:13, 1:12, 1:11, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 1:0.5, 1:0.4, 1:0.3, 1:0.2, or 1:0.1, or any range between any of the foregoing values, such as between 1:1 and 1:10 or 1:0.2 to 1:12, each comprise an endpoint. In some embodiments, the plurality of ratios includes any or all of the ratios provided herein. In some embodiments, the recombinant receptor stimulant is a cell that expresses an antigen recognized by a recombinant receptor. In some embodiments, the recombinant receptor is a CAR, and the stepwise adjusted number of cells of the therapeutic cell composition are incubated with a constant number of such particles at a plurality of ratios of cells to particles of the therapeutic cell composition, such as at or about 1:100, 1:75, 1:50, 1:40, 1:30, 1:20, 1:15, 1:14, 1:13, 1:12, 1:11, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 1:0.5, 1:0.4, 1:0.3, 1:0.2, or 1:0.1, or a range between any of the foregoing values, such as a ratio between 1:1 and 1:10 or 1:0.2 to 1:12, each comprising an end value. In some embodiments, the plurality of ratios includes any or all of the ratios provided herein.
In some embodiments, the recombinant receptor stimulant is a cell that expresses a target (e.g., antigen or antibody) recognized by the recombinant receptor. In some embodiments, a constant number of cells of a recombinant receptor (e.g., CAR) and therapeutic cell composition are incubated with the cells at a plurality of ratios of cells of a therapeutic cell composition to cells expressing a target (e.g., antigen or antibody) including at or about 1:100, 1:75, 1:50, 1:40, 1:30, 1:20, 1:15, 1:14, 1:13, 1:12, 1:11, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 1:0.5, 1:0.4, 1:0.3, 1:0.2, or 1:0.1, or a range between any of the foregoing values, such as a ratio between 1:1 and 1:10 or 1:0.2 to 1:12, each comprising an endpoint. In some embodiments, the plurality of ratios includes any or all of the ratios provided herein. In some embodiments, the recombinant receptor is a CAR, and the stepwise adjusted number of cells of the therapeutic cell composition are incubated with a constant number of cells expressing the target (e.g., antigen or antibody) at a plurality of ratios of cells of the therapeutic cell composition to cells expressing the antigen, including at or about 1:100, 1:75, 1:50, 1:40, 1:30, 1:20, 1:15, 1:14, 1:13, 1:12, 1:11, 1:10, 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 1:0.5, 1:0.4, 1:0.3, 1:0.2, or 1:0.1, or a range between any of the foregoing values, such as a ratio between 1:1 and 1:10 or 1:0.2 to 1:12, each comprising an endpoint. In some embodiments, the plurality of ratios includes any or all of the ratios provided herein.
In some embodiments, the cell composition is administered at about 1x10 2 And about 1x10 4 Between each of about 1x10 3 And about 1x10 5 Between each of about 1x10 4 And about 1x10 6 Between each of about 1x10 5 And about 1x10 7 Between each of about 1x10 6 And about 1x10 8 Between each of about 1x10 7 And about 1x10 9 Between each, and about 1x10 8 And about 1x10 10 Cells between each (each containing an endpoint) are incubated with a constant amount or concentration of recombinant receptor stimulant.
In some embodiments, cells of the therapeutic cell composition are incubated with the recombinant receptor stimulant in a volume of cell culture medium. It should be understood that the exact volume may be determined empirically and vary with the surface area of the vessel (e.g., multiwell plate) in which the assay is performed. In certain embodiments, the cells are incubated with the recombinant receptor stimulating agent in a volume of at least or about 1 μl, at least or about 10 μl, at least or about 25 μl, at least or about 50 μl, at least or about 100 μl, at least or about 500 μl, at least or about 1mL, at least or about 1.5mL, at least or about 2mL, at least or about 2.5mL, at least or about 5mL, at least or about 10mL, at least or about 20mL, at least or about 25mL, at least or about 50mL, at least or about 100mL, or greater than 100 mL. In certain embodiments, the cells are incubated with the recombinant receptor stimulant in a volume that falls between about 1 μl and about 100 μl, between about 100 μl and about 500 μl, between about 500 μl and about 1mL, between about 1mL and about 10mL, between about 10mL and about 50mL, or between about 10mL and about 100mL (each inclusive). In certain embodiments, the cells are incubated with the recombinant receptor stimulant in a volume of between about 100 μl and about 1mL (inclusive). In a particular embodiment, the cells are incubated with the recombinant receptor stimulant in a volume of about 500 μl. In some embodiments, the multi-well plate is a 6-well plate and the volume is from or about 1mL to or about 3mL. In some embodiments, the multi-well plate is a 12-well plate and the volume is from or about 1mL to or about 2mL. In some embodiments, the multi-well plate is a 24-well plate and the volume is from or about 0.5mL to or about 1mL. In some embodiments, the multi-well plate is a 48-well plate and the volume is from or about 0.2mL to or about 0.4mL. In some embodiments, the multiwell plate is a 96-well plate and the volume is from or about 0.1mL to or about 0.2mL.
In some embodiments, a constant number of cells of the therapeutic cell composition are incubated with a recombinant receptor stimulator at a concentration of between about 1fmol and about 1pmol, between about 1pmol and about 1nmol, between about 1nmol and about 1 μmol, between about 1 μmol and about 1mmol, or between about 1mmol and 1mol (each comprising an end value). In particular embodiments, a constant number of cells of the therapeutic cell composition are incubated with a recombinant receptor stimulator at a concentration of between about 1fM and about 1pM, between about 1pM and about 1nM, between about 1nM and about 1 μM, between about 1 μM and about 1mM, or between about 1mM and 1mol (each comprising an endpoint). Exemplary units include, but are not limited to, pg/mL, pg/(mL/hr), pg (mL x cell), pg/(mL x hr x cell), and pg/(mL x hr x 10) 6 Individual cells).
In certain embodiments, for each of the plurality of ratios tested, the measure of recombinant receptor-dependent activity (e.g., CAR-dependent activity) is the amount or concentration, or relative amount or concentration, of the soluble factor in the therapeutic cell composition at a time point during or at the end of the incubation period. In particular embodiments, the measurement is subtracted from the control measurement or normalized to the control measurement. In some embodiments, the control measurement is a measurement from the same cell composition taken prior to incubation. In certain embodiments, the control measurement is a measurement taken from the same control cell composition that has not been incubated with the binding molecule. In certain embodiments, the control is a measurement taken from a cell composition that does not contain recombinant receptor positive cells at the same time point during incubation with the binding molecule.
In some embodiments, the measurement is a normalized ratio of amount or concentration as compared to a control. In a particular embodiment, the measurement is a per amount of time (e.g.,an amount or concentration of the soluble factor per minute or hour). In some embodiments, the measurement is per cell or per set or reference number of cells (e.g., per 100 cells, per 10 3 Every 10 cells 4 Every 10 cells 5 Every 10 cells 6 Individual cells, etc.). In certain embodiments, the measurement is an amount or concentration of the soluble factor per amount of time, per cell, or per reference number of cells. In some embodiments, the measurement is the amount or concentration of soluble factor per cell expressing the recombinant receptor. In certain embodiments, the measurement is the amount or concentration of the soluble factor per amount of time (e.g., per minute or hour) per cell expressing the recombinant receptor (car+ cell) of the therapeutic cell composition. In some embodiments, the measurement is the amount or concentration of the soluble factor per amount or concentration of recombinant receptor or recombinant receptor stimulant per amount of time. In some embodiments, the measurement is of the amount or concentration of the soluble factor per cell or per set or reference number of cells per amount or concentration of recombinant receptor stimulant. In some embodiments, the measurement is the amount or concentration of soluble factor per amount of time, per recombinant receptor or recombinant receptor stimulator, per cell or per reference number of cells. In some embodiments, the measurement is the amount or concentration of soluble factor per recombinant receptor or recombinant receptor stimulator, per cell expressing the recombinant receptor. In certain embodiments, the measurement is the amount or concentration of soluble factor per amount of time, per amount or concentration of recombinant receptor or recombinant receptor stimulant, per amount of car+ cells of the therapeutic cell composition.
In particular embodiments, the recombinant receptor or CAR-dependent activity is the production or secretion of two or more soluble factors. In certain embodiments, the recombinant receptor-dependent activity or CAR-dependent activity is the production or secretion of two, three, four, five, six, seven, eight, nine, ten, or more than ten soluble factors. In some embodiments, the measured values of two, three, four, five, six, seven, eight, nine, ten, or more than ten soluble factors are combined into an arithmetic average or geometric average. In certain measurements, the measure of recombinant receptor-dependent activity is the secretion or complexation value of two, three, four, five, six, seven, eight, nine, ten, or more than ten soluble factors.
In certain embodiments, the measurement of the recombinant receptor-dependent activity is transformed, for example, by a logarithmic transformation. In certain embodiments, the measure of recombinant receptor activity is measured by common log (log 10 (x) Natural logarithm (ln (x)) or binary logarithm (log) 2 (x) A) transform. In some embodiments, the measure of recombinant receptor-dependent activity is a composite of measures of production or secretion of two or more soluble factors. In some embodiments, two or more measurements of the production or secretion of soluble factors are transformed prior to combining into a composite measurement. In certain embodiments, the measurement of the recombinant receptor-dependent activity is transformed prior to normalization relative to a reference measurement. In certain embodiments, the measurement of the recombinant receptor-dependent activity is transformed prior to normalization relative to a reference measurement. In some embodiments, normalization of the recombinant receptor-dependent activity is performed relative to the maximum recombinant receptor-dependent activity measured from multiple incubations.
In certain embodiments, the soluble factor is a cytokine. Cytokines are a large group of small signaling molecules that play a broad role in cellular communication. Cytokines are most often associated with a variety of immunomodulatory molecules, including interleukins, chemokines and interferons. Alternatively, cytokines can be characterized by their structure, which is classified into four families: a four alpha helix family comprising the IL-2 subfamily, the IFN subfamily, and the IL-10 subfamily; IL-1 family, IL-17 family, and cysteine knot cytokines including transforming growth factor beta family members. In some embodiments, the recombinant receptor-dependent activity is the production or secretion of one or more soluble factors including interleukins, interferons and chemokines. In particular embodiments, the recombinant receptor-dependent activity (e.g., CAR-dependent activity) is an IL-2 family member, an IFN subfamily member, an IL-10 subfamily member; production or secretion of one or more of an IL-1 family member, an IL-17 family member, a cysteine knot cytokine, and/or a member of the transforming growth factor beta family.
In particular embodiments, the recombinant receptor-dependent activity or CAR-dependent activity is the production and/or secretion of one or more of IL-1, IL-1 β, IL-2, sIL-2Ra, IL-3, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12, IL-13, IL-27, IL-33, IL-35, TNF, tnfα, CXCL2, CCL3, CCL5, CCL17, CCL24, PGD2, LTB4, interferon γ (IFN- γ), granulocyte macrophage colony-stimulating factor (GM-CSF), macrophage Inflammatory Protein (MIP) -1a, MIP-1b, flt-3L, fractal chemokine, and/or IL-5. In certain embodiments, the CAR-dependent activity is the production or secretion of a Th17 cytokine. In some embodiments, the Th17 cytokine is GMCSF. In some embodiments, the CAR dependent activity comprises the production or secretion of a Th2 cytokine, wherein the Th2 cytokine is IL-4, IL-5, IL-10 or IL-13.
In certain embodiments, the recombinant receptor-dependent activity or CAR-dependent activity is the production or secretion of a proinflammatory cytokine. Pro-inflammatory cytokines play a role in initiating inflammatory responses and regulate host defenses against pathogens that mediate innate immune responses. Proinflammatory cytokines include, but are not limited to, interleukin (IL), interleukin-l-beta (IL-1), interleukin-3 (IL-3), interleukin-5 (IL-5), interleukin-6 (IL-6), interleukin-13 (IL-13), tumor Necrosis Factor (TNF), CXC-chemokine ligand 2 (CXCL 2), CC-chemokine ligand 2 (CCL 2), CC-chemokine ligand 3 (CCL 3), CC-chemokine ligand 5 (CCL 5), CC-chemokine ligand 17 (CCL 17), CC-chemokine ligand 24 (CCL 24), prostaglandin D2 (PGD 2), and leukotriene B4 (LTB 4), and IL-33. In some embodiments, the recombinant receptor-dependent activity or CAR-dependent activity is the production and/or secretion of interleukins and/or TNF family members. In particular embodiments, the recombinant receptor-dependent activity or CAR-dependent activity is the production and/or secretion of IL-1, IL-6, IL-8, and IL-18, TNF- α, or a combination thereof.
In particular embodiments, the recombinant receptor activity (e.g., CAR-dependent activity) is secretion of IL-2, IFN- γ, TNF- α, or a combination thereof. In some embodiments, the recombinant receptor activity (e.g., CAR-dependent activity) is secretion of IL-2. In some embodiments, the recombinant receptor activity (e.g., CAR-dependent activity) is secretion of IFN- γ. In some embodiments, the recombinant receptor activity (e.g., CAR-dependent activity) is secretion of TNF- α.
In particular embodiments, the recombinant receptor-dependent activity is the cytolytic (cytotoxic) activity of the therapeutic cell composition. In some embodiments, recombinant receptor-dependent cytolytic activity is assessed by: exposing a cell expressing a recombinant receptor or a cell composition comprising a cell expressing a recombinant receptor to a varying amount of a target cell expressing an antigen and/or epitope bound and/or recognized by the recombinant receptor, incubating with a varying amount of a target cell expressing an antigen and/or epitope bound and/or recognized by the recombinant receptor, and/or contacting with a varying amount of a target cell expressing an antigen and/or epitope bound and/or recognized by the recombinant receptor. Cytolytic activity may be measured by directly or indirectly measuring the number of target cells over time. For example, the target cells may be incubated with a detectable label (such a label that is detectable and then the target cells are lysed, or a detectable label that is detectable in the presence of the target cells) prior to incubation with the recombinant receptor expressing cells. These readings provide direct or indirect target cell numbers and/or target cell death, and can be measured at different time points during the assay. A decrease in the number of target cells and/or an increase in target cell death is indicative of cytolytic activity of the cells. Suitable methods for performing a cytolytic assay are known in the art and include, but are not limited to, chromium-51 release assays, non-radioactive chromium assays, flow cytometry assays using fluorescent dyes such as carboxyfluorescein succinimidyl ester (CFSE), PKH-2, and PKH-26.
In certain embodiments, recombinant receptor (e.g., CAR) -dependent cytolytic activity is measured by incubating a cell composition comprising cells expressing the recombinant receptor with a target cell expressing an antigen or epitope thereof bound or recognized by the recombinant receptor. In certain embodiments, the recombinant receptor is a CAR. In some embodiments, cells of the therapeutic cell composition are incubated with cells expressing antigen in a ratio comprising a ratio (each comprising an end value) of 10:1, about 5:1, about 4:1, about 3:1, about 2:1, about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, or about 1:10, or between 10:1 and 1:1, between 3:1 and 1:3, or between 1:1 and 1:10. In some embodiments, cells of the cell composition are incubated with target cells at a ratio of car+ cells of the therapeutic cell composition to target cells that includes a ratio of about 10:1, about 5:1, about 4:1, about 3:1, about 2:1, about 1:1, about 1:2, about 1:3, about 1:4, about 1:5, about 1:6, about 1:7, about 1:8, about 1:9, or about 1:10, or a ratio between 10:1 and 1:1, between 3:1 and 1:3, or between 1:1 and 1:10 (each comprising an end value).
In certain embodiments, the cells of the therapeutic cell composition are incubated with the target cells for up to or about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 8 hours, about 12 hours, about 18 hours, about 24 hours, about 48 hours, or greater than 48 hours. In some embodiments, a constant number of cells of the therapeutic cell composition are incubated with the antigen-expressing cells for about 18 hours, 19 hours, 20 hours, 21 hours, 22 hours, 23 hours, or 24 hours. In some embodiments, the therapeutic cell composition is administered at about 1x10 2 And about 1x10 4 Between each of about 1x10 3 And about 1x10 5 Between each of about 1x10 4 And about 1x10 6 Between each of about 1x10 5 And about 1x10 7 Between each of about 1x10 6 And about 1x10 8 Between each of about 1x10 7 And about 1x10 9 Between each, or at about 1x10 8 And about 1x10 10 Cells between each (each containing an end value) are incubated with varying numbers of antigen expressing cells to generate a plurality of ratios. In certain embodiments, therapeutic cells are combinedAt about 1x10 2 And about 1x10 4 Between each of about 1x10 3 And about 1x10 5 Between each of about 1x10 4 And about 1x10 6 Between each of about 1x10 5 And about 1x10 7 Between each of about 1x10 6 And about 1x10 8 Between each of about 1x10 7 And about 1x10 9 Between each, or at about 1x10 8 And about 1x10 10 A constant amount of car+ cells (each containing an end value) between each is incubated with varying numbers of antigen expressing cells to generate multiple ratios.
In some embodiments, the measure of activity is compared to a control. In certain embodiments, the control is a culture of antigen expressing cells that have not been incubated with the cell composition. In some embodiments, the control is a measurement from a control cell composition that does not contain car+ cells incubated with antigen-expressing cells at the same ratio.
In certain embodiments, for each ratio tested, the measure of cytolytic activity assay is the number of antigen-expressing cells that survive at a time point during or at the end of the incubation. In certain embodiments, the measurement is the amount of a target cell death marker (e.g., chromium-51) released during incubation. In some embodiments, the measurement is the amount of target cell death determined by subtracting the amount of target cells in co-incubation at a given time point from the amount of target cells of the control incubated alone. In some embodiments, the measured value is the percentage of target cells that are maintained at a time point compared to the starting amount of target cells. In certain embodiments, the measured value is the amount of cells killed within a certain amount of time. In certain embodiments, the measured value is the amount of killed cells per cell in the cell composition. In some embodiments, the measurement is the amount of killed cells per cell, or per set or reference number of cells, such as, but not limited to, per 100 cells, per 10 cells of the composition 3 Every 10 cells 4 Every 10 cells 5 Every 10 cells 6 Every 10 cells 7 Every 10 cells 8 Every 10 cells 9 Individual cells, or every 10 10 Amount of target cells killed by individual cells. In a particular embodiment, the measurement is the amount of each car+ cell or a reference or set number of killed cells thereof in the cell composition. In certain embodiments, the measured value is the amount of killed cells per cell of the cell composition over a period of time. In a particular embodiment, the measurement is the amount of killed cells per car+ cell of the therapeutic cell composition over a certain amount of time.
In some embodiments, the recombinant receptor-dependent activity is upregulation of a gene in a cell of the therapeutic cell composition. In some embodiments, recombinant receptor-dependent gene upregulation activity is assessed by: exposing a cell expressing a recombinant receptor or a cell composition comprising a cell expressing a recombinant receptor to, incubating with, and/or contacting with a varying amount of a recombinant receptor stimulator that binds to and stimulates a recombinant receptor. Upregulation of gene activity may be measured by direct or indirect amounts over time.
In some embodiments, the recombinant receptor-dependent activity is down-regulation of a gene in a cell of the therapeutic cell composition. In some embodiments, recombinant receptor-dependent gene downregulating activity is assessed by: exposing a cell expressing a recombinant receptor or a cell composition comprising a cell expressing a recombinant receptor to, incubating with, and/or contacting with a varying amount of a recombinant receptor stimulator that binds to and stimulates a recombinant receptor. Down-regulation of gene activity may be measured by direct or indirect amounts over time.
In some embodiments, the recombinant receptor-dependent activity is upregulation of the receptor in the cells of the therapeutic cell composition. In some embodiments, recombinant receptor-dependent receptor up-regulation activity is assessed by: exposing a cell expressing a recombinant receptor or a cell composition comprising a cell expressing a recombinant receptor to, incubating with, and/or contacting with a varying amount of a recombinant receptor stimulator that binds to and stimulates a recombinant receptor. Up-regulation of receptor activity can be measured by direct or indirect amounts over time.
In some embodiments, the recombinant receptor-dependent activity is down-regulation of a receptor in a cell of the therapeutic cell composition. In some embodiments, recombinant receptor-dependent receptor downregulating activity is assessed by: exposing a cell expressing a recombinant receptor or a cell composition comprising a cell expressing a recombinant receptor to, incubating with, and/or contacting with a varying amount of a recombinant receptor stimulator that binds to and stimulates a recombinant receptor. Downregulation of receptor activity may be measured by direct or indirect amounts over time.
In some embodiments, the measured values of the recombinant receptor-dependent activity are fitted using a mathematical model to generate a recombinant receptor-dependent activity curve. In some cases, curve fitting may allow for the inference or extrapolation of the behavior of therapeutic cell compositions, such as recombinant receptor-dependent activity. It is contemplated that any method known in the art may be used to perform curve fitting. In some embodiments, the curve is S-shaped. In some embodiments, the stepwise adjustment ratio resulting in half-maximal recombinant receptor-dependent activity is determined based on recombinant receptor-dependent activity measured from each of the plurality of incubations. In some embodiments, the stepwise adjustment ratio resulting in half-maximal recombinant receptor-dependent activity is extrapolated, or estimated from a recombinant receptor-dependent activity curve. In some embodiments, the recombinant receptor-dependent activity curve is normalized to the measured maximum recombinant receptor-dependent activity. In some embodiments, the recombinant receptor-dependent activity curve is normalized to a series of values of the higher asymptote of the curve, optionally the higher asymptote.
In some embodiments, methods comprising assays as described herein can be performed in duplicate or in triplicate or more to verify measurements of recombinant receptor-dependent activity. In some cases, such as in duplicate, triplicate, or more assays, the measured recombinant receptor-dependent activity from each replicate experiment is used to provide a descriptive statistical measure of the recombinant receptor-dependent activity. For example, in some cases, for each of the plurality of ratios tested, an average (e.g., arithmetic average), median, standard deviation, and/or variance of each measure of recombinant receptor-dependent activity is determined. In some embodiments, the average value of each measure of recombinant receptor-dependent activity is determined. In some embodiments, the standard deviation of each measure of recombinant receptor-dependent activity is determined. In some embodiments, the average measure of recombinant receptor-dependent activity is fitted using a mathematical model to generate or estimate a recombinant receptor-dependent activity curve. In some embodiments, the curve is normalized to the average maximum. In some embodiments, the curve is normalized to the average of a series of values of the higher asymptote, optionally the higher asymptote.
The metrics described herein may be used with respect to a reference standard, such as the reference standard described herein (e.g., section I-D-1).
D. Determining efficacy of therapeutic cell compositions
The methods provided herein allow for determining the efficacy of a therapeutic cell composition. It is contemplated that the assays described herein can be used to assess the efficacy of therapeutic cell compositions made by processes such as those described herein (e.g., section II) as well as any other manufacturing process that allows cells of the therapeutic cell composition made to be cultured in an assay comprising a plurality of incubations, wherein each incubation comprises incubating cells of the therapeutic composition in a different ratio with a recombinant receptor stimulator capable of stimulating recombinant receptor-dependent activity of the therapeutic cell composition. In some embodiments, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or more therapeutic cell compositions can be evaluated according to the methods provided herein.
By taking measurements of recombinant receptor-dependent activity at each of the plurality of ratios tested, the efficacy of therapeutic cell compositions can be determined. In some embodiments, the measurement is a composite value determined by taking an arithmetic mean or median value in duplicate, triplicate, or more replicates. In some embodiments, the standard deviation and/or variance of the measurements may be determined. In some embodiments, one or more measurements (including composite measurements) of the recombinant receptor-dependent activity of a therapeutic cell composition in response to a recombinant receptor stimulator can be used to determine the efficacy of the therapeutic cell composition. In some embodiments, the recombinant receptor-dependent activity may be any one as described in section I-C. In some embodiments, the recombinant receptor stimulant may be any one as described in section I-B.
In some embodiments, multiple incubations at different ratios produce multiple measurements that can be applied to a curve fitting method. In some embodiments, the plurality of measurements includes a composite measurement (e.g., average or median). For example, the recombinant receptor-dependent activity measurements can be fitted with a curve (e.g., sigmoid) to allow for inference, extrapolation, or estimation of the behavior (e.g., sensitivity) of the therapeutic cell composition. In some embodiments, a curve fitted to the measurements may be used to estimate the behavior (e.g., sensitivity) of the therapeutic composition that was not directly examined during the assay. For example, a curve may be used to estimate a lower asymptote; a minimum value; loss of detection of recombinant receptor-dependent activity; a specified percentage of maximum (e.g., 10%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, or 90%); half maximum (e.g., 50% recombinant receptor-dependent activity); a range of 10% -90%, 20% -80%, 30% -70% or 40% -60% of the maximum recombinant receptor-dependent activity (i.e., the maximum activity as described below); a higher asymptote; and a maximum value and a ratio at which each of the values or ranges occurs.
It is contemplated that any measure (ratio at half maximum, range, maximum, minimum, asymptote, and composite measures thereof) may be used to determine the efficacy of the therapeutic cell composition. In some embodiments, the potency is a relative potency.
1. Efficacy of
In some embodiments, the efficacy of a therapeutic cell composition is defined as the ratio at which one or more or a series of recombinant receptor-dependent activity measurements occur. In some embodiments, one or more or a series of measurements are composite measurements, such as an average or median value determined from repeated experiments. In some embodiments, the measurement and ratio are determined from a recombinant receptor-dependent activity curve of the measured recombinant receptor-dependent activity. In some embodiments, the measured recombinant receptor-dependent activity is normalized to the maximum activity measured for the therapeutic composition. In some embodiments, the recombinant receptor-dependent activity curve is normalized to the maximum recombinant receptor-dependent activity measured for the therapeutic cell composition. In some embodiments, the recombinant receptor-dependent activity curve is normalized relative to a higher asymptote for the recombinant receptor-dependent activity measured for the therapeutic cell composition, optionally across the average of the measurements of the asymptote.
In some embodiments, the efficacy of a therapeutic cell composition is a ratio range within which 10% -90% recombinant receptor-dependent activity occurs, or vice versa. In some embodiments, the ratio range (within which 10% -90% of the recombinant receptor-dependent activity occurs) is estimated from a recombinant receptor-dependent activity curve. In some embodiments, for example when the recombinant receptor-dependent activity measure or the recombinant receptor-dependent activity curve is normalized, the range of recombinant receptor-dependent activity values is 0.1-0.9 or 10% -90%.
In some embodiments, the efficacy of a therapeutic cell composition is a ratio range within which 20% -80% recombinant receptor-dependent activity occurs, or vice versa. In some embodiments, the ratio range (within which 20% -80% of the recombinant receptor-dependent activity occurs) is estimated from a recombinant receptor-dependent activity curve. In some embodiments, for example when the recombinant receptor-dependent activity measure or the recombinant receptor-dependent activity curve is normalized, the range of recombinant receptor-dependent activity values is 0.2-0.8 or 20% -80%.
In some embodiments, the efficacy of a therapeutic cell composition is a ratio range within which 30% -70% recombinant receptor-dependent activity occurs, or vice versa. In some embodiments, the ratio range (within which 30% -70% of the recombinant receptor-dependent activity occurs) is estimated from a recombinant receptor-dependent activity curve. In some embodiments, for example when the recombinant receptor-dependent activity measure or the recombinant receptor-dependent activity curve is normalized, the range of recombinant receptor-dependent activity values is 0.3-0.7 or 30% -70%.
In some embodiments, the efficacy of a therapeutic cell composition is a ratio range within which 40% -60% recombinant receptor-dependent activity occurs, or vice versa. In some embodiments, the ratio range (within which 40% -60% of the recombinant receptor-dependent activity occurs) is estimated from a recombinant receptor-dependent activity curve. In some embodiments, for example when the recombinant receptor-dependent activity measure or the recombinant receptor-dependent activity curve is normalized, the range of recombinant receptor-dependent activity values is 0.4-0.6 or 40% -60%.
In some embodiments, the potency of a therapeutic cell composition is the ratio at which half maximal recombinant receptor-dependent activity occurs. In some embodiments, the half-maximum and the ratio (at which half-maximum occurs) are estimated from a recombinant receptor-dependent activity curve. In some embodiments, for example, when the recombinant receptor-dependent activity measure or the recombinant receptor-dependent activity curve is normalized, the half maximum recombinant receptor-dependent activity value is 0.5 or 50%.
In some embodiments, for example when the recombinant receptor-dependent activity curve is fitted by sigmoid, the linear portion of the curve is determined. In some embodiments, the efficacy is a measurement from the linear portion of the curve and the corresponding ratio. In some embodiments, the half maximum measurement and ratio are determined from the linear portion of the curve.
2. Relative efficacy
The methods provided herein allow for determining the efficacy of a therapeutic cell composition relative to a different therapeutic cell composition (e.g., a reference standard). This type of efficacy may be referred to as relative efficacy. For example, therapeutic cell compositions assessed according to the methods provided herein can be compared to different therapeutic cell compositions (e.g., reference standards, such as described below) assessed according to the methods provided herein, for example, to determine how the efficacy of the therapeutic cell compositions correlate with each other. This provides an advantage in that multiple therapeutic cell compositions can be compared to determine which composition has the highest or best efficacy. In some embodiments, the optimal efficacy is one that can elicit a therapeutic effect (e.g., sustained response, progression free survival) in the subject. In some embodiments, the optimal efficacy is one that does not result in toxicity in the subject. In some embodiments, the optimal efficacy is one that can elicit a therapeutic effect (e.g., sustained response, progression free survival) in the subject and that does not result in toxicity.
In some embodiments, the relative potency of a therapeutic cell composition is defined as comparing one or more ratios (one or more or a series of recombinant receptor-dependent activity measurements occurring at the one or more ratios) for the therapeutic cell composition to one or more ratios (one or more or a series of recombinant receptor-dependent activity measurements occurring at the one or more ratios) for a reference standard. In some embodiments, one or more or a series of measurements of one or both of the therapeutic cell composition and the reference standard is a composite measurement, such as an average or median value determined from repeated experiments. In some embodiments, the measured values and ratios of the therapeutic cell composition and the reference standard are determined from a recombinant receptor-dependent curve of the measured recombinant receptor-dependent activity of the composition, respectively. In some embodiments, the measured recombinant receptor-dependent activity of the therapeutic cell composition and the reference standard is normalized to the maximum activity measured for the therapeutic composition and the reference standard, respectively. In some embodiments, the recombinant receptor-dependent activity curves of the therapeutic cell composition and the reference standard are normalized relative to the maximum recombinant receptor-dependent activity measured for the therapeutic cell composition and the reference standard, respectively. In some embodiments, the recombinant receptor-dependent activity curves of the therapeutic cell composition and the reference standard are normalized relative to the higher asymptote of the recombinant receptor-dependent activity measured for the therapeutic cell composition and the reference standard, respectively, optionally across the average of the measurements of the asymptote.
In some embodiments, the relative potency of a therapeutic cell composition is compared to a range of ratios (10% -90% recombinant receptor-dependent activity occurs within the range of ratios, or vice versa) to a range for a reference standard (10% -90% recombinant receptor-dependent activity occurs within the range, or vice versa). In some embodiments, the ratio range for the therapeutic cell composition and the reference standard at which 10% -90% of the recombinant receptor-dependent activity occurs is estimated from the recombinant receptor-dependent activity curves of the therapeutic cell composition and the reference standard, respectively. In some embodiments, for example, when the measure of recombinant receptor-dependent activity or the recombinant receptor-dependent activity curve of the therapeutic cell composition and the reference standard is normalized, the range of recombinant receptor-dependent activity values is 0.1-0.9 or 10% -90%.
In some embodiments, the relative potency of a therapeutic cell composition is compared to a range of ratios (20% -80% recombinant receptor-dependent activity occurs within the range of ratios, or vice versa) to a range for a reference standard (20% -80% recombinant receptor-dependent activity occurs within the range, or vice versa). In some embodiments, the ratio range for the therapeutic cell composition and the reference standard at which 20% -80% of the recombinant receptor-dependent activity occurs is estimated from the recombinant receptor-dependent activity curves of the therapeutic cell composition and the reference standard, respectively. In some embodiments, for example, when the measure of recombinant receptor-dependent activity or the recombinant receptor-dependent activity curve of the therapeutic cell composition and the reference standard is normalized, the range of recombinant receptor-dependent activity values is 0.2-0.8 or 20% -80%.
In some embodiments, the relative potency of a therapeutic cell composition is compared to a range of ratios (30% -70% recombinant receptor-dependent activity occurs within the range of ratios, or vice versa) to a range for a reference standard (30% -70% recombinant receptor-dependent activity occurs within the range, or vice versa). In some embodiments, the ratio range for the therapeutic cell composition and the reference standard at which 30% -70% of the recombinant receptor-dependent activity occurs is estimated from the recombinant receptor-dependent activity curves of the therapeutic cell composition and the reference standard, respectively. In some embodiments, for example, when the measure of recombinant receptor-dependent activity or the recombinant receptor-dependent activity curve of the therapeutic cell composition and the reference standard is normalized, the range of recombinant receptor-dependent activity values is 0.3-0.7 or 30% -70%.
In some embodiments, the relative potency of a therapeutic cell composition is compared to a range of ratios (40% -60% recombinant receptor-dependent activity occurs within the range of ratios, or vice versa) to a range for a reference standard (40% -60% recombinant receptor-dependent activity occurs within the range, or vice versa). In some embodiments, the ratio range for the therapeutic cell composition and the reference standard at which 40% -60% of the recombinant receptor-dependent activity occurs is estimated from the recombinant receptor-dependent activity curves of the therapeutic cell composition and the reference standard, respectively. In some embodiments, for example, when the measure of recombinant receptor-dependent activity or the recombinant receptor-dependent activity curve of the therapeutic cell composition and the reference standard is normalized, the range of recombinant receptor-dependent activity values is 0.4-0.6 or 40% -60%.
In some embodiments, the relative potency of a therapeutic cell composition is a ratio at which half maximal recombinant receptor-dependent activity occurs compared to a ratio to a reference standard at which half maximal recombinant receptor-dependent activity occurs. In some embodiments, the half-maximum and ratio for the therapeutic cell composition and the reference standard (at which half-maximum occurs) are estimated from recombinant receptor-dependent activity curves of the therapeutic cell composition and the reference standard, respectively. In some embodiments, for example, when the measure of recombinant receptor-dependent activity or the recombinant receptor-dependent activity curve of the therapeutic cell composition and the reference standard is normalized, the half-maximal recombinant receptor-dependent activity value is 0.5 or 50%.
In some embodiments, for example, when the recombinant receptor-dependent activity profile of the therapeutic cell composition and the reference standard are fitted by sigmoid, the linear portion of the profile is determined. In some embodiments, the relative efficacy is a comparison of the measured value and corresponding ratio from the linear portion of the curve of the therapeutic cell composition to the measured value and corresponding ratio from the linear portion of the curve of the reference standard. In some embodiments, the half-maximum measurement and ratio of therapeutic cell composition to reference standard is determined from the linear portion of the curve.
In some embodiments, the comparison between the measured values of the therapeutic cell composition and the reference composition (as described above) is a division. For example, the ratio for a therapeutic cell composition at which half maximal recombinant receptor-dependent activity occurs is divided by the ratio for a reference standard at which half maximal recombinant receptor-dependent activity occurs. In some embodiments, the relative efficacy is expressed as a ratio. In some embodiments, the relative efficacy is expressed as a percentage.
In some embodiments, for example, when the recombinant receptor-dependent activity curves of the therapeutic cell composition and the reference standard are fitted by sigmoid and normalized as described above, the relative potency is the difference between the curves. In some embodiments, the difference between the curves is measured for the linear portion of the normalized curve. In some embodiments, normalization of the recombinant receptor-dependent activity curves (e.g., sigmoid curves) of the therapeutic cell composition and the reference standard can be used to directly compare the recombinant receptor-dependent activity curves of the therapeutic cell composition and the reference standard.
a. Reference standard
Particular embodiments contemplate that a measurement of recombinant receptor-dependent activity (e.g., car+ dependent activity) of a therapeutic cell composition can be compared to a reference measurement (i.e., reference measure) of a reference standard, for example, to determine relative potency. In certain embodiments, the reference measurement is a predetermined measurement of the recombinant receptor-dependent activity of a reference standard or a value thereof. In some embodiments, the recombinant receptor-dependent activity of a reference standard is assessed according to the methods disclosed herein. In some embodiments, the reference standard is a therapeutic cell composition that has been validated to result in a stepwise adjustment of the ratio of recombinant receptor-dependent activity. In some embodiments, the reference standard is a therapeutic cell composition that has been validated for a stepwise adjustment ratio that results in recombinant receptor-dependent activity, and the measured activity has been curve (e.g., sigmoid) fitted to generate a recombinant receptor-dependent activity curve. In some embodiments, the recombinant receptor-dependent activity curve of the reference standard is normalized. In some embodiments, the recombinant receptor-dependent activity curve is normalized to the maximum measured recombinant receptor-dependent activity. In some embodiments, the recombinant receptor-dependent activity curve is normalized relative to the higher asymptote of the recombinant receptor-dependent activity curve. In some embodiments, the recombinant receptor-dependent activity curve is normalized relative to an average calculated on the higher asymptote of the recombinant receptor-dependent activity curve. In some embodiments, the reference standard is a therapeutic cell composition comprising a validated step-by-step adjustment ratio that results in half-maximal recombinant receptor-dependent activity. In some embodiments, the validated step-by-step adjustment ratio that results in half-maximal recombinant receptor-dependent activity is determined from a recombinant receptor-dependent activity curve.
In some embodiments, the reference standard is a commercially available therapeutic cell composition. In some embodiments, the reference standard is a therapeutic cell composition manufactured using the same manufacturing process as the therapeutic cell composition used for the manufacture and comparison thereof. In some embodiments, the reference standard is a therapeutic cell composition manufactured using a manufacturing process that is different from the manufacturing process used to manufacture the therapeutic cell composition with which it is compared. In some embodiments, the reference standard is a therapeutic cell composition comprising the same recombinant receptor as the therapeutic cell composition to which it is compared. In some embodiments, the reference standard is a therapeutic cell composition comprising a different recombinant receptor than the therapeutic cell composition to which it is compared. In some embodiments, the reference standard is a therapeutic cell composition that is manufactured from the same subject as the therapeutic cell composition to which it is compared. In some embodiments, the reference standard is a therapeutic cell composition that is manufactured from a different subject than the therapeutic cell composition to which it is compared. In some embodiments, the reference standard is a therapeutic cell composition derived from a healthy subject. In some embodiments, the reference standard is derived from a subject suffering from a disease or disorder. In some embodiments, the reference standard is derived from a subject having cancer. In some embodiments, the reference standard may be a combination of one or more of those described above.
In some embodiments, a reference standard has been administered to a subject. In certain embodiments, administration of the reference standard to the subject is observed and determined to result in an acceptable safety profile upon administration to the subject. In certain embodiments, administration of the reference standard does not result in any serious toxicity. In certain embodiments, administration of the reference standard does not result in any serious neurotoxicity. In particular embodiments, the reference standard is a therapeutic cell composition associated with a grade 4 or lower, grade 3 or lower, grade 2 or lower, grade 1 or lower, or grade 0 scored neurotoxicity. In some embodiments, the reference standard is associated with an acceptable safety feature. In particular embodiments, an acceptable safety feature is the absence of observed grade 1 or higher, observed grade 2 or higher, observed grade 3 or higher, or grade 4 or higher neurotoxicity. In certain embodiments, the reference standard is associated with acceptable safety profiles in the absence of observed grade 3 or higher neurotoxicity. In certain embodiments, the reference standard is associated with acceptable safety profiles in the absence of observed grade 3 or higher neurotoxicity.
In certain embodiments, it has been observed or determined that the reference standard results in a desired efficacy upon administration to a subject. In certain embodiments, the subject has a disease or disorder that expresses an antigen or is associated with an antigen, as is the subject to which the reference standard is administered. In certain embodiments, it has been observed or determined that a reference standard results in a Complete Response (CR). In certain embodiments, it has been observed or determined that a reference standard results in a sustained response. Methods for generating engineered T cells
In some embodiments, the methods of potency of the therapeutic cell compositions provided herein can be used in combination with therapeutic compositions (e.g., export compositions) that produce engineered cells (e.g., engineered cd4+ T cells and/or engineered cd8+ T cells) that express recombinant proteins, e.g., recombinant receptors (e.g., T Cell Receptors (TCRs) or Chimeric Antigen Receptors (CARs)). In some embodiments, the processes provided herein are used in conjunction with the manufacture, generation, or generation of cell therapies, and may be used in conjunction with additional processing steps, such as isolation, separation, selection, activation, or stimulation, transduction, washing, suspending, diluting, concentrating, and/or formulating cells. In some embodiments, the process of generating or producing an engineered cell, e.g., an engineered cd4+ T cell and/or an engineered cd8+ T cell, comprises one or more of the following: isolating cells from a subject, preparing cells, processing cells, incubating cells under stimulating conditions, and/or engineering (e.g., transducing) cells. In some embodiments, the process comprises processing steps performed in the following order: first isolating, e.g., selecting or separating, input cells, e.g., primary cells, from a biological sample; incubating an input cell under stimulating conditions, the input cell being engineered with a vector particle, e.g., a viral vector particle, to introduce a recombinant polynucleotide into the cell, e.g., by transduction or transfection; incubating the engineered cells (e.g., transduced cells), such as to expand the cells; and collecting, harvesting all or a portion of the cells, and/or filling the container with the same to formulate the cells into an output composition. In some embodiments, cd4+ and cd8+ T cells are manufactured independently of each other, e.g., in separate input compositions, but the manufacturing process includes the same processing steps. In some embodiments, the cd4+ and cd8+ T cells are manufactured together, for example, in the same input composition. In some embodiments, cells of the produced output composition (e.g., therapeutic cell composition) are reintroduced into the same subject either before or after cryopreservation. In some embodiments, the engineered cell export composition (e.g., therapeutic cell composition) is suitable for use in therapy (e.g., autologous cell therapy, allogeneic cell therapy). An exemplary manufacturing process is described in published international patent application publication No. WO 2019/089855, the contents of which are incorporated herein by reference in their entirety.
A. Sample and cell preparation
In particular embodiments, the provided methods are used in combination with one or more input compositions that separate, select, and/or enrich cells from a biological sample to generate enriched cells (e.g., T cells). In some embodiments, provided methods include isolating cells or a composition thereof from a biological sample, such as those obtained from or derived from a subject, such as a subject having a particular disease or disorder or in need of cell therapy or to be administered cell therapy. In some aspects, the subject is a human, such as a subject that is a patient in need of a particular therapeutic intervention (e.g., adoptive cell therapy in which cells are isolated, processed, and/or engineered for use in the adoptive cell therapy). Thus, in some embodiments, the cell is a primary cell, such as a primary human cell. Samples include tissues, fluids, and other samples taken directly from a subject. The biological sample may be a sample obtained directly from a biological source or a processed sample. Biological samples include, but are not limited to, body fluid (e.g., blood, plasma, serum, cerebrospinal fluid, synovial fluid, urine, and sweat), tissue and organ samples, including samples derived from their processing.
In some aspects, the sample is a blood or blood-derived sample, or is derived from apheresis or a leukocyte apheresis product. Exemplary samples include whole blood, peripheral Blood Mononuclear Cells (PBMCs), leukocytes, bone marrow, thymus, tissue biopsies, tumors, leukemias, lymphomas, lymph nodes, intestinal-related lymphoid tissue, mucosa-related lymphoid tissue, spleen, other lymphoid tissue, liver, lung, stomach, intestine, colon, kidney, pancreas, breast, bone, prostate, cervix, testes, ovaries, tonsils, or other organs and/or cells derived therefrom. In the case of cell therapies (e.g., adoptive cell therapies), the samples include samples from autologous and allogeneic sources.
In some examples, cells from the circulating blood of the subject are obtained, for example, by apheresis or leukocyte apheresis. In some aspects, the sample contains lymphocytes (including T cells, monocytes, granulocytes, B cells), other nucleated leukocytes, erythrocytes, and/or platelets, and in some aspects contains cells other than erythrocytes and platelets.
In some embodiments, blood cells collected from a subject are washed, e.g., to remove plasma fractions, and the cells are placed in an appropriate buffer or medium for subsequent processing steps. In some embodiments, the cells are washed with Phosphate Buffered Saline (PBS). In some embodiments, the wash solution lacks calcium and/or magnesium and/or many or all divalent cations. In some aspects, the flow is controlled by a semi-automatic "flow-through" centrifuge (e.g. Cobe 2991 cell processor, baxter) completed the washing step. In some aspects, the washing step is accomplished by Tangential Flow Filtration (TFF) according to manufacturer's instructions. In some embodiments, cells are resuspended in a plurality of biocompatible buffers (e.g., such as Ca-free ++ /Mg ++ PBS of (x). In certain embodiments, components of the blood cell sample are removed and the cells are resuspended directly in culture medium.
In some embodiments, the method of preparation comprises the step of freezing (e.g., cryopreserving) the cells before or after isolation, selection and/or enrichment, and/or incubation for transduction and engineering, and/or after incubating and/or harvesting the engineered cells. In some embodiments, the freezing and subsequent thawing steps remove granulocytes and, to some extent, monocytes from the cell population. In some embodiments, the cells are suspended in a chilled solution to remove plasma and platelets, for example, after a washing step. In some aspects, any of a variety of known freezing solutions and parameters may be used. In some embodiments, the cells are frozen, e.g., cryogenically frozen or cryogenically preserved, in a medium and/or solution having a final concentration of DMSO of or about 12.5%, 12.0%, 11.5%, 11.0%, 10.5%, 10.0%, 9.5%, 9.0%, 8.5%, 8.0%, 7.5%, 7.0%, 6.5%, 6.0%, 5.5%, or 5.0%, or DMSO of between 1% and 15%, between 6% and 12%, between 5% and 10%, or between 6% and 8%. In particular embodiments, the cells are frozen, e.g., cryogenically frozen or cryogenically preserved, in a medium and/or solution having a final concentration of HSA of or about 5.0%, 4.5%, 4.0%, 3.5%, 3.0%, 2.5%, 2.0%, 1.5%, 1.25%, 1.0%, 0.75%, 0.5% or 0.25%, or between 0.1% and-5%, between 0.25% and 4%, between 0.5% and 2%, or between 1% and 2%. One example includes using PBS containing 20% DMSO and 8% Human Serum Albumin (HSA), or other suitable cell freezing medium. It was then diluted 1:1 with medium such that the final concentrations of DMSO and HSA were 10% and 4%, respectively. The cells are then typically frozen at a rate of at or about 1 °/minute to at or about-80 ℃ and stored in the gas phase of a liquid nitrogen storage tank.
In some embodiments, the isolation of the cells or populations includes one or more preparative and/or non-affinity based cell isolation steps. In some examples, cells are washed, centrifuged, and/or incubated in the presence of one or more reagents, e.g., to remove unwanted components, enrich for desired components, lyse, or remove cells sensitive to a particular reagent. In some examples, cells are isolated based on one or more characteristics, such as density, adhesion characteristics, size, sensitivity to a particular component, and/or resistance to a drug. In some embodiments, the methods include density-based cell separation methods, such as the preparation of leukocytes from peripheral blood by lysing erythrocytes and centrifuging through a Percoll or Ficoll gradient.
In some embodiments, at least a portion of the selecting step comprises incubating the cells with a selection agent. Incubation with one or more selection reagents, for example, as part of a selection method, may be performed using one or more selection reagents for selecting one or more different cell types based on the expression or presence of one or more specific molecules, such as surface markers (e.g., surface proteins), intracellular markers, or nucleic acids, in or on the cell. In some embodiments, any known method of separation based on such labels using one or more selection reagents may be used. In some embodiments, the one or more selection reagents result in an isolation that is affinity-based or immunoaffinity-based. For example, in some aspects, selection includes incubation with one or more reagents for isolating cells and cell populations based on cell expression or expression levels of one or more markers (typically cell surface markers), e.g., by incubation with antibodies or binding partners that specifically bind to such markers, followed by a washing step and isolating cells that have bound to the antibodies or binding partners from those that have not bound to the antibodies or binding partners.
In some aspects of such processes, a volume of cells is mixed with a quantity of an affinity-based desired selection reagent. Immunoaffinity-based selection can be performed using any system or method that allows for an advantageous energy interaction between the isolated cells and a labeled molecule that specifically binds to the cells (e.g., an antibody or other binding partner on a solid surface (e.g., a particle)). In some embodiments, the method is performed using particles, such as beads (e.g., magnetic beads), coated with a selective agent (e.g., an antibody) that is specific for a marker of the cell. Particles (e.g., beads) may be incubated or mixed with cells in a container (e.g., tube or bag) while shaking or mixing, wherein the ratio of cell density to particles (e.g., beads) is constant to help promote energetically favorable interactions. In other cases, the method comprises selecting cells, wherein all or a portion of the selection is performed in an internal cavity of a centrifugal chamber, e.g., under centrifugal rotation. In some embodiments, incubating the cells with a selective agent (e.g., a selective agent based on immunoaffinity) is performed in the centrifugal chamber. In certain embodiments, the separation or isolation is performed using a system, apparatus or device described in international patent application publication No. WO 2009/072003 or US20110003380 A1. In one example, the system is a system as described in International publication No. WO 2016/073602.
In some embodiments, by implementing such selection steps or portions thereof (e.g., incubation with antibody-coated particles (e.g., magnetic beads)) in the cavity of the centrifuge chamber, the user is able to control certain parameters, such as the volume of the various solutions, the addition of solutions during processing, and timing thereof, which may provide a number of advantages over other available processes. For example, the ability to reduce the volume of liquid in the chamber during incubation can increase the concentration of particles (e.g., bead reagents) used in the selection, thereby increasing the chemical potential of the solution without affecting the total number of cells in the chamber. This in turn may enhance the pairwise interactions between the cells being processed and the particles for selection. In some embodiments, for example, when associated with systems, circuits, and controls as described herein, an incubation step is performed in the chamber, allowing the user to achieve agitation of the solution at one or more desired times during incubation, which may also improve interaction.
In some embodiments, at least a portion of the selecting step is performed in a centrifugal chamber, which includes incubating the cells with a selection agent. In some aspects of such processes, a volume of cells is mixed with a desired amount of affinity-based selection reagent that is significantly less than the volume and amount typically used when similar selections are made in a tube or container to select the same number of cells and/or the same volume of cells according to manufacturer's instructions. In some embodiments, the amount of the one or more selection reagents employed is no more than 5%, no more than 10%, no more than 15%, no more than 20%, no more than 25%, no more than 50%, no more than 60%, no more than 70%, or no more than 80% of the amount of the same one or more selection reagents used to select cells for the same number of cells and/or the same volume of cells in a tube or container-based incubation according to manufacturer's instructions.
In some embodiments, for selection of cells, e.g., based on immunoaffinity selection, the cells are incubated in a chamber cavity in a composition that also contains a selection buffer with a selection reagent, e.g., a surface-labeled molecule, such as an antibody, that specifically binds to cells that are desired to be enriched and/or depleted (but not to other cells in the composition), optionally coupled to a scaffold (e.g., a polymer or surface, e.g., a bead, e.g., a magnetic bead, such as a magnetic bead coupled to monoclonal antibodies specific for CD4 and CD 8). In some embodiments, as described, the selection reagent is added to the cells in the chamber cavity in an amount that is generally used or will require significantly less (e.g., no more than 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, or 80%) of the amount of selection reagent that would be used to achieve about the same or similar selection efficiency for the same number of cells or the same volume of cells when selected in an oscillating or rotating tube. In some embodiments, incubation is performed with the addition of a selection buffer to the cells and selection reagent to achieve a target volume of incubation of, for example, 10mL to 200mL, such as at least or about 10mL, 20mL, 30mL, 40mL, 50mL, 60mL, 70mL, 80mL, 90mL, 100mL, 150mL, or 200mL of reagent. In some embodiments, the selection buffer and the selection reagent are pre-mixed prior to addition to the cells. In some embodiments, the selection buffer and the selection reagent are added separately to the cells. In some embodiments, the selective incubation is performed under periodically mild mixing conditions, which may help promote energetically favorable interactions, allowing for the use of less total selection reagent while achieving high selection efficiency.
In some embodiments, the total duration of incubation with the selection agent is from 5 minutes to 6 hours or from about 5 minutes to about 6 hours, such as 30 minutes to 3 hours, for example at least or about at least 30 minutes, 60 minutes, 120 minutes, or 180 minutes.
In some embodiments, incubation is typically performed under mixing conditions, such as in the presence of rotation, typically at a relatively low force or speed, such as a speed less than the speed used to precipitate the cells, such as from 600rpm to 1700rpm or from about 600rpm to about 1700rpm (e.g., at or about or at least 600rpm, 1000rpm, or 1500rpm, or 1700 rpm), such as at a RCF of from 80g to 100g or from about 80g to about 100g (e.g., at or about or at least 80g, 85g, 90g, 95g, or 100 g) of the sample or wall of the chamber or other container. In some embodiments, the rotation is performed using a repeating interval of such low speed rotation and a subsequent rest period, such as rotation and/or rest for 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 seconds, such as about 1 or 2 seconds, followed by rest for about 5, 6, 7, or 8 seconds.
In some embodiments, this process is performed in a completely closed system integral to the chamber. In some embodiments, this process (and in some aspects one or more additional steps, such as a pre-wash step to wash the cell-containing sample, such as a apheresis sample) is performed in an automated manner such that cells, reagents, and other components are aspirated and pushed out of the chamber at the appropriate times and centrifuged to complete the washing and binding steps in a single closed system using an automated procedure.
In some embodiments, after incubating and/or mixing the cells with one or more selection reagents, the incubated cells are isolated to select the cells based on the presence or absence of the one or more specific reagents. In some embodiments, the separation is performed in the same closed system, wherein the cells are incubated with the selection agent. In some embodiments, after incubation with the selection reagent, the incubated cells (including cells in which the selection reagent has been bound) are transferred into the system for separation of cells based on immunoaffinity. In some embodiments, the system for immunoaffinity-based separation is or contains a magnetic separation column.
Such isolation steps may be based on positive selection (where cells that have bound an agent (e.g., an antibody or binding partner) are retained for further use) and/or negative selection (where cells that have not bound an agent (e.g., an antibody or binding partner) are retained). In some examples, both fractions are retained for further use. In some aspects, negative selection may be particularly useful in the absence of antibodies useful for specifically identifying cell types in heterogeneous populations, such that isolation is preferably based on markers expressed by cells other than the desired population.
In some embodiments, the process step further comprises negative and/or positive selection of the incubated cells, e.g., using a system or apparatus that can perform affinity-based selection. In some embodiments, the separation is performed by enriching a specific cell population via positive selection, or depleting a specific cell population via negative selection. In some embodiments, positive or negative selection is accomplished by: incubating the cells with one or more antibodies or other binding agents that specifically bind to one or more surface markers that are expressed on positively or negatively selected cells (markers+) or at relatively high levels (markers, respectively High height ). Multiple rounds of the same selection step (e.g., positive or negative selection steps) may be performed. At the position ofIn certain embodiments, the positive or negative selected fraction is subjected to a selection process, such as by repeating the positive or negative selection step. In some embodiments, the selection is repeated twice, three times, four times, five times, six times, seven times, eight times, nine times, or more than nine times. In certain embodiments, the same selection is performed up to five times. In certain embodiments, the same selection step is performed three times.
Isolation need not result in 100% enrichment or removal of a particular cell population or cells expressing a particular marker. For example, positive selection or enrichment of cells of a particular type (such as those expressing a marker) refers to increasing the number or percentage of such cells, but without the need to have cells that do not express the marker completely absent. Likewise, negative selection, removal, or depletion of particular types of cells (such as those expressing a marker) refers to reducing the number or percentage of such cells, but does not require complete removal of all such cells.
In some examples, multiple rounds of separation steps are performed, wherein fractions from positive or negative selection of one step are subjected to another separation step, such as subsequent positive or negative selection. In some examples, a single isolation step may deplete cells expressing multiple markers simultaneously, such as by incubating the cells with multiple antibodies or binding partners (each antibody or binding partner being specific for a marker targeted for negative selection). Likewise, by incubating the cells with multiple antibodies or binding partners expressed on various cell types, positive selection can be performed on multiple cell types simultaneously. In certain embodiments, one or more separation steps are repeated and/or performed more than once. In some embodiments, the positive or negative selection fractions resulting from the separation step are subjected to the same separation step, such as by repeating the positive or negative selection step. In some embodiments, a single isolation step is repeated and/or performed more than once, e.g., to increase the yield of positively selected cells, to increase the purity of negatively selected cells, and/or to further remove positively selected cells from a negatively selected fraction. In certain embodiments, one or more separation steps are performed and/or repeated two, three, four, five, six, seven, eight, nine, ten, or more than ten times. In certain embodiments, one or more selection steps are performed and/or repeated between one and ten times, between one and five times, or between three and five times. In certain embodiments, one or more selection steps are repeated three times.
For example, in some aspects, specific subsets of T cells are isolated by positive or negative selection techniques, such as cells positive for one or more surface markers or expressing high levels of surface markers, e.g., cd28+, cd62l+, CCR7+, cd27+, cd127+, cd4+, cd8+, cd45ra+ and/or cd45ro+ T cells. In some embodiments, such cells are selected by incubation with one or more antibodies or binding partners that specifically bind such labels. In some embodiments, the antibody or binding partner may be conjugated (e.g., directly or indirectly) to a solid support or matrix (e.g., magnetic beads or paramagnetic beads) to effect selection. For example, CD3+, CD28+ T cells can be generated using CD3/CD28 conjugated magnetic beads (e.g.,m-450 CD3/CD 28T cell expander and/or +.>Beads) were positively selected.
In some embodiments, T cells are isolated from a PBMC sample by negative selection for a marker expressed on non-T cells (e.g., B cells, monocytes, or other leukocytes such as CD 14). In some aspects, a cd4+ or cd8+ selection step is used to isolate cd4+ helper T cells and cd8+ cytotoxic T cells. Such cd4+ and cd8+ populations may be further sorted into sub-populations by positive or negative selection for markers expressed or expressed to a relatively high degree on one or more naive T cells, memory T cells, and/or effector T cell sub-populations.
In some embodiments, the cd8+ T cells are further enriched or depleted for naive, central memory, effector memory, and/or central memory stem cells, such as by positive or negative selection based on surface antigens associated with the respective subpopulations. In some embodiments, enrichment is performed for central memory T (TCM) cells to increase efficacy, such as to improve long-term survival, expansion, and/or transplantation after administration, which is particularly robust in some aspects in such subpopulations. See Terakura et al, (2012) blood.1:72-82; wang et al (2012) J Immunother.35 (9): 689-701. In some embodiments, combining TCM-enriched cd8+ T cells with cd4+ T cells further enhances efficacy.
In embodiments, memory T cells are present in both cd62l+ and CD 62L-subsets of cd8+ peripheral blood lymphocytes. PBMCs may be enriched or depleted against CD62L-cd8+ and/or cd62l+cd8+ fractions, e.g. using anti-CD 8 and anti-CD 62L antibodies.
In some embodiments, enrichment of central memory T (TCM) cells is based on positive or high surface expression of CD45RO, CD62L, CCR7, CD28, CD3 and/or CD 127; in some aspects, it is based on negative selection of cells expressing or highly expressing CD45RA and/or granzyme B. In some aspects, isolation of a cd8+ population enriched for TCM cells is performed by depletion of cells expressing CD4, CD14, CD45RA and positive selection or enrichment of cells expressing CD 62L. In one aspect, enrichment of central memory T (TCM) cells is performed starting from a negative cell fraction selected based on CD4 expression, which is subjected to negative selection based on expression of CD14 and CD45RA and positive selection based on CD 62L.
In some aspects this selection is made simultaneously, while in other aspects it is made sequentially in either order. In some aspects, the same CD4 expression-based selection step used to prepare the population or subpopulation of cd8+ T cells is also used to generate the population or subpopulation of cd4+ T cells such that both positive and negative fractions from the CD 4-based isolation are retained and used in subsequent steps of the method, optionally after one or more other positive or negative selection steps. In some embodiments, the selection of the cd4+ T cell population and the selection of the cd8+ T cell population are performed simultaneously. In some embodiments, the selection of the cd4+ T cell population and the cd8+ T cell population is performed sequentially in either order. In some embodiments, the methods for selecting cells may include those described in published U.S. application No. US 20170037369. In some embodiments, the selected cd4+ T cell population and the selected cd8+ T cell population may be combined after selection. In some aspects, the selected cd4+ T cell population and the selected cd8+ T cell population may be combined in a bioreactor bag as described herein. In some embodiments, the population of selected cd4+ T cells and the population of selected cd8+ T cells are processed separately, as according to the provided methods, thereby enriching the population of selected cd4+ T cells for cd4+ T cells and incubating them with a stimulating agent (e.g., anti-CD 3/anti-CD 28 magnetic beads), transduced with a viral vector encoding a recombinant protein (e.g., CAR) and incubated under conditions that expand T cells; and enriching the selected population of cd8+ T cells for cd8+ T cells and incubating them with a stimulating agent (e.g., anti-CD 3/anti-CD 28 magnetic beads), transduced with a viral vector encoding a recombinant protein (e.g., CAR) such as the same recombinant protein used to engineer cd4+ T cells from the same donor, and incubated under conditions to expand T cells.
In certain embodiments, a biological sample (e.g., a sample of PBMCs or other leukocytes) is subjected to selection of cd4+ T cells, wherein both negative and positive fractions are retained. In certain embodiments, the cd8+ T cells are selected from the negative fraction. In some embodiments, the biological sample is subjected to selection of cd8+ T cells, wherein both negative and positive fractions are retained. In certain embodiments, the cd4+ T cells are selected from the negative fraction.
In a specific example, a PBMC sample or other leukocyte sample is subjected to selection of cd4+ T cells, wherein both negative and positive fractions are retained. The negative fractions are then negative for the expression of CD14 and CD45RA or CD19 and positive for the marker characteristics of central memory T cells (such as CD62L or CCR 7), in any order.
By identifying a population of cells with cell surface antigens, cd4+ T helper cells can be sorted into naive, central memory and effector cells. Cd4+ lymphocytes can be obtained by standard methods. In some embodiments, the naive cd4+ T lymphocytes are cd45ro-, cd45ra+, cd62l+, or cd4+ T cells. In some embodiments, the central memory cd4+ T cells are cd62l+ and cd45ro+. In some embodiments, effector CD4+ T cells are CD 62L-and CD45RO-.
In one example, to enrich for cd4+ T cells by negative selection, a monoclonal antibody cocktail typically includes antibodies to CD14, CD20, CD11b, CD16, HLA-DR, and CD 8. In some embodiments, the antibody or binding partner is bound to a solid support or matrix (e.g., magnetic or paramagnetic beads) to allow separation of cells for positive and/or negative selection. For example, in some embodiments immunomagnetic (or affinity magnetic) separation techniques are used to separate or isolate cells and cell populations (reviewed in Methods in Molecular Medicine, volume 58: metastasis Research Protocols, volume 2: cell Behavior In Vitro and In Vivo, pages 17-25 S.A.Brooks and U.S. Schumacher edition)Humana Press Inc.,Totowa,NJ)。
In some aspects, the incubated cell sample or composition to be separated is combined with a magnetic device containing a small magnetizable or magnetically responsive material (e.g., magnetically responsive particles or microparticles, such as paramagnetic beads (e.g., such as Dynabeads or DynabeadsBeads) are incubated with the selection reagent. The magnetically responsive material (e.g., particles) is typically directly or indirectly attached to a binding partner (e.g., an antibody) that specifically binds to a molecule (e.g., a surface label) present on a cell, cells, or cell population that is desired to be isolated (e.g., desired to be selected negatively or positively).
In some embodiments, the magnetic particles or beads comprise magnetically responsive material that binds to a specific binding member (e.g., an antibody or other binding partner). Many well-known magnetically responsive materials for use in magnetic separation methods are known, such as those described in U.S. Pat. No. 4,452,773 to Molday and European patent Specification EP 452342B, which are hereby incorporated by reference. Colloid-sized particles may also be used, such as those described in U.S. patent No. 4,795,698 to Owen; and those described in us patent No. 5,200,084 to Liberti et al.
Incubation is typically performed under conditions whereby the antibody or binding partner, or a molecule that specifically binds to such antibody or binding partner attached to a magnetic particle or bead (e.g., a secondary antibody or other agent), specifically binds to a cell surface molecule (if present on cells within the sample).
In certain embodiments, the magnetically responsive particles are coated in a primary or other binding partner, secondary antibody, lectin, enzyme or streptavidin. In certain embodiments, the magnetic particles are attached to the cells by coating with a primary antibody specific for one or more markers. In certain embodiments, cells are labeled with a primary antibody or binding partner instead of beads, and then magnetic particles coated with a cell type specific secondary antibody or other binding partner (e.g., streptavidin) are added. In certain embodiments, streptavidin-coated magnetic particles are used in combination with biotinylated primary or secondary antibodies.
In some aspects, separation is achieved in a procedure in which the sample is placed in a magnetic field, and those cells having magnetically responsive or magnetizable particles attached thereto will be attracted to the magnet and separated from unlabeled cells. For positive selection, cells attracted by the magnet are retained; for negative selection, cells that were not attracted (unlabeled cells) were retained. In some aspects, a combination of positive and negative selections is performed during the same selection step, wherein the positive and negative fractions are retained and further processed or subjected to additional separation steps.
In some embodiments, affinity-based selection is performed by Magnetic Activated Cell Sorting (MACS) (Miltenyi Biotech, obu, ca). Magnetically Activated Cell Sorting (MACS) (e.g., a clinic MACS system) enables high purity selection of cells having magnetized particles attached thereto. In certain embodiments, MACS operates in a mode in which non-target and target species are eluted sequentially after application of an external magnetic field. That is, cells attached to the magnetized particles remain in place, while unattached species are eluted. Then, after the first elution step is completed, the species that are trapped in the magnetic field and prevented from eluting are released in some way so that they can be eluted and recovered. In certain embodiments, non-target cells are labeled and depleted from a heterogeneous cell population.
In some embodiments, the magnetically responsive particles remain attached to cells that are subsequently incubated, cultured, and/or engineered; in some aspects, the particles remain attached to the cells for administration to a patient. In some embodiments, the magnetizable or magnetically responsive particles are removed from the cells. Methods for removing magnetizable particles from cells are known and include, for example, the use of competitive non-labeled antibodies, magnetizable particles or antibodies conjugated with cleavable linkers, and the like. In some embodiments, the magnetizable particles are biodegradable.
In some embodiments, one or more import compositions that produce enriched T cells, e.g., cd3+ T cells, cd4+ T cells, and/or cd8+ T cells, are isolated and/or selected. In some embodiments, two or more separate input compositions are isolated, selected, enriched, or obtained from a single biological sample. In some embodiments, the separate input compositions are isolated, selected, enriched, and/or obtained from separate biological samples collected, obtained, and/or obtained from the same subject.
In certain embodiments, one or more input compositions are or include a composition enriched for T cells comprising at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or at or about 100% cd3+ T cells. In a particular embodiment, the T cell enriched input composition consists essentially of cd3+ T cells.
In certain embodiments, one or more input compositions are or include a composition enriched for cd4+ T cells comprising at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or at or about 100% cd4+ T cells. In certain embodiments, the input composition of cd4+ T cells comprises less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1%, or less than 0.01% cd8+ T cells, and/or is free of cd8+ T cells, and/or is free or substantially free of cd8+ T cells. In some embodiments, the T cell enriched composition consists essentially of cd4+ T cells.
In certain embodiments, one or more compositions are or comprise cd8+ T cells, which are or comprise at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or at or about 100% cd8+ T cells. In certain embodiments, the composition of cd8+ T cells contains less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1%, or less than 0.01% of cd4+ T cells, and/or does not contain or is substantially free of cd4+ T cells. In some embodiments, the T cell enriched composition consists essentially of cd8+ T cells.
In some embodiments, one or more of the input compositions enriched for T cells are frozen after isolation, selection and/or enrichment, e.g., cryopreserved and/or cryogenically frozen. In some embodiments, the one or more input compositions are frozen, e.g., cryopreserved and/or cryogenically frozen, prior to any step of incubating, activating, stimulating, engineering, transducing, transfecting, incubating, amplifying, harvesting, and/or formulating the composition of the cells. In particular embodiments, one or more cryogenically frozen input compositions are stored, for example, at or about-80 ℃ for 12 hours to 7 days, 24 hours to 120 hours, or 2 days to 5 days. In particular embodiments, one or more cryogenically frozen input compositions are stored at or about-80 ℃ for an amount of time less than 10 days, 9 days, 8 days, 7 days, 6 days or 5 days, 4 days, 3 days, 2 days or 1 day. In some embodiments, one or more cryogenically frozen input compositions are stored or stored at about-80 ℃ for about 1, 2, 3, 4, 5, or 6 days.
B. Activation and stimulation of cells
In some embodiments, the provided methods are used in combination with incubating the cells under stimulating conditions. In some embodiments, the stimulation conditions include activating or stimulating and/or conditions capable of activating or stimulating a signal (e.g., a signal generated from a TCR and/or co-receptor) in a cell (e.g., a cd4+ T cell or a cd8+ T cell). In some embodiments, the stimulation conditions include one or more steps of culturing, incubating, activating, propagating the cells with a stimulating agent (e.g., an agent that activates or stimulates and/or is capable of activating or stimulating a signal in the cells) and/or in the presence of a stimulating agent. In some embodiments, the stimulating agent stimulates and/or activates the TCR and/or co-receptor. In certain embodiments, the stimulating agent is an agent as described in section II-B-1.
In certain embodiments, one or more compositions enriched for T cells are incubated under stimulating conditions prior to genetically engineering the cells, such as by transfection and/or transduction of the cells by the techniques provided in section II-C. In certain embodiments, after one or more compositions of enriched T cells have been isolated, selected, enriched, or obtained from a biological sample, the one or more compositions are incubated under stimulating conditions. In certain embodiments, one or more of the compositions is an input composition. In certain embodiments, one or more of the input compositions have been previously cryogenically frozen and stored and thawed prior to incubation.
In certain embodiments, the one or more compositions enriched for T cells are or include two separate compositions, e.g., separate input compositions, for the enriched T cells. In particular embodiments, two separate compositions of enriched T cells, e.g., two separate compositions of enriched T cells selected, isolated, and/or enriched from the same biological sample, are incubated under stimulation conditions, respectively. In certain embodiments, the two separate compositions comprise a composition enriched for cd4+ T cells. In a particular embodiment, the two separate compositions comprise a composition enriched for cd8+ T cells. In some embodiments, the two separate compositions enriched for cd4+ T cells and enriched for cd8+ T cells are incubated under stimulation conditions, respectively.
In some embodiments, a single composition enriched for T cells is incubated under stimulating conditions. In certain embodiments, the single composition is a composition enriched for cd4+ T cells. In some embodiments, the single composition is a composition enriched for cd4+ and cd8+ T cells that have been combined from separate compositions prior to incubation.
In some embodiments, the composition enriched for cd4+ T cells incubated under stimulation conditions comprises at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or is about 100% cd4+ T cells. In certain embodiments, the composition of enriched cd4+ T cells incubated under stimulation conditions comprises less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1% or less than 0.01% cd8+ T cells, and/or is free of cd8+ T cells, and/or is free or substantially free of cd8+ T cells.
In some embodiments, the composition enriched for cd8+ T cells incubated under stimulation conditions comprises at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or at or about 100% cd8+ T cells. In certain embodiments, the composition of enriched cd8+ T cells incubated under stimulation conditions comprises less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1%, or less than 0.01% of cd4+ T cells, and/or is free or substantially free of cd4+ T cells.
In some embodiments, separate compositions enriched for cd4+ and cd8+ T cells are combined into a single composition and incubated under stimulating conditions. In certain embodiments, the separate stimulated compositions of enriched cd4+ and enriched cd8+ T cells are combined into a single composition after incubation has been performed and/or completed. In some embodiments, as in accordance with the provided methods, the separately stimulated compositions of stimulated cd4+ T and stimulated cd8+ T cells are processed separately after incubation has been performed and/or completed, whereby a population of stimulated cd4+ T cells (e.g., incubated with a stimulating anti-CD 3/anti-CD 28 magnetic bead stimulating agent) is transduced with a viral vector encoding a recombinant protein (e.g., CAR) and incubated under conditions that expand T cells, and a population of stimulated cd8+ T cells (e.g., incubated with a stimulating anti-CD 3/anti-CD 28 magnetic bead stimulating agent) is transduced with a viral vector encoding a recombinant protein (e.g., CAR) (e.g., the same recombinant protein as used to engineer cd4+ T cells from the same donor) and incubated under conditions that expand T cells.
In some embodiments, incubating under stimulation conditions may include culturing, incubating, stimulating, activating, propagating, including by incubating in the presence of stimulation conditions, e.g., conditions designed to induce proliferation, expansion, activation, and/or survival of cells in the population, mimicking antigen exposure, and/or priming cells for genetic engineering (e.g., for introduction of recombinant antigen receptors). In particular embodiments, the stimulation conditions may include one or more of the following: specific media, temperature, oxygen content, carbon dioxide content, time, agents (e.g., nutrients, amino acids, antibiotics, ions, and/or stimulatory factors (e.g., cytokines, chemokines, antigens, binding partners, fusion proteins, recombinant soluble receptors, and any other agents designed to activate cells)).
In some aspects, the stimulation and/or incubation under stimulation conditions is performed according to a variety of techniques, such as those described in the following documents: U.S. Pat. No. 6,040,1,77 to Riddell et al; klebaroff et al (2012) J Immunother.35 (9): 651-660; terakura et al (2012) blood.1:72-82; and/or Wang et al (2012) J Immunother35 (9): 689-701.
In some embodiments, cells (e.g., T cells), cell compositions, and methods are expanded byAnd/or cell populations, e.g. CD4 + And CD8 + T cells or a composition, population or subpopulation thereof: adding feeder cells, such as non-dividing Peripheral Blood Mononuclear Cells (PBMCs), to the culture starting composition (e.g., such that the resulting cell population contains at least about 5, 10, 20, or 40 or more PBMC feeder cells for each T lymphocyte in the initial population to be expanded); and incubating the culture (e.g., for a time sufficient to expand the number of T cells). In some aspects, the non-dividing feeder cells may comprise gamma irradiated PBMC feeder cells. In some embodiments, PBMCs are irradiated with gamma rays ranging from about 3000 to 3600 rads to prevent cell division. In some aspects, feeder cells are added to the medium prior to the addition of the T cell population.
In some embodiments, the stimulation conditions include a temperature suitable for growth of human T lymphocytes, for example, at least about 25 degrees celsius, typically at least about 30 degrees celsius, and typically at or about 37 degrees celsius. In some embodiments, the temperature transition is achieved during the incubation, such as from 37 degrees celsius to 35 degrees celsius. Optionally, the incubation may further comprise adding non-dividing EBV transformed Lymphoblastoid Cells (LCLs) as feeder cells. The LCL may be irradiated with gamma rays in the range of about 6000 to 10,000 rads. In some aspects, the LCL feeder cells are provided in any suitable amount (e.g., a ratio of LCL feeder cells to naive T lymphocytes of at least about 10:1).
In embodiments, antigen-specific CD4 may be obtained by stimulating naive or antigen-specific T lymphocytes with an antigen + And CD8 + Is a group of the above-mentioned groups. For example, antigen-specific T cell lines or clones can be generated against cytomegalovirus antigens by isolating T cells from an infected subject and stimulating the cells in vitro with the same antigen. Naive T cells may also be used.
In particular embodiments, the stimulating conditions include incubating, culturing, and/or incubating the cells with a stimulating agent. In certain embodiments, the stimulating agent is an agent as described in section II-B-1. In certain embodiments, the stimulating agent comprises or includes a bead. Exemplary stimulating agents are or include anti-CD 3/anti-CD 28 magnetic beads. In certain embodiments, when the cells are contacted with and/or incubated with a stimulating agent, the incubation, culturing, and/or initiation of culturing the cells under stimulating conditions occurs. In certain embodiments, the cells are incubated before, during, and/or after genetically engineering the cells (e.g., introducing the recombinant polynucleotide into the cells, such as by transfection or transduction).
In some embodiments, the T cell enriched composition is incubated with the cells at or at a ratio of about 3:1, 2.5:1, 2:1, 1.5:1, 1.25:1, 1.2:1, 1.1:1, 1:1, 0.9:1, 0.8:1, 0.75:1, 0.67:1, 0.5:1, 0.3:1, or 0.2:1 stimulating agent and/or beads (e.g., anti-CD 3/anti-CD 28 magnetic beads). In particular embodiments, the ratio of stimulating agent and/or bead to cells is between 2.5:1 and 0.2:1, between 2:1 and 0.5:1, between 1.5:1 and 0.75:1, between 1.25:1 and 0.8:1, between 1.1:1 and 0.9:1. In particular embodiments, the ratio of stimulating agent to cells is about 1:1 or 1:1.
In certain embodiments, incubating the cells at a ratio (e.g., a ratio of 1:1) of less than 3:1 or less than 3 stimulating agents (e.g., anti-CD 3/anti-CD 28 magnetic beads) per cell reduces the amount of cell death (e.g., as a result of activation-induced cell death) that occurs during incubation. In some embodiments, cells are incubated with a stimulating agent (e.g., anti-CD 3/anti-CD 28 magnetic beads) at a bead to cell ratio of less than 3 (or 3:1 or less than 3 beads per cell). In certain embodiments, incubating the cells at a ratio of less than 3:1 or less than 3 beads per cell (e.g., a ratio of 1:1) reduces the amount of cell death (e.g., as a result of activation-induced death) that occurs during incubation.
In particular embodiments, the T cell enriched composition is incubated with a stimulating agent (e.g., anti-CD 3/anti-CD 28 magnetic beads) at a stimulating agent and/or bead to cell ratio of less than 3:1 (e.g., a ratio of 1:1), and at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99% or at least 99.9% of the T cells survive, e.g., are viable and/or do not undergo necrosis, programmed cell death or apoptosis, for a period of 1, 2, 3, 4, 5, 6, 7, or more than 7 days or at least 1, 2, 3, 4, 5, 6, 7, or more than 7 days after the incubation is completed. In particular embodiments, the T cell enriched composition is incubated with a stimulating agent at a stimulating agent and/or bead to cell ratio of less than 3:1 (e.g., a 1:1 ratio), and less than 50%, less than 40%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1%, or less than 0.01% of the cells undergo activation-induced cell death during incubation.
In certain embodiments, the T cell enriched composition is incubated with a stimulating agent (e.g., anti-CD 3/anti-CD 28 magnetic beads) at a ratio of bead to cell of less than 3:1 (e.g., a ratio of 1:1), and the cells of the composition have a viability that is at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 100%, at least 150%, at least 1 fold, at least 2 fold, at least 3 fold, at least 4 fold, at least 5 fold, at least 10 fold, at least 25 fold, at least 50 fold, or at least 100 fold greater than cells that have undergone an exemplary and/or alternative procedure in which the T cell enriched composition is incubated with a stimulating agent at a ratio of 3:1 or greater.
In some embodiments, the composition of enriched T cells incubated with the stimulating agent comprises from 1.0x10 5 Individual cells/mL to 1.0x10 8 Individual cells/mL or from about 1.0x10 5 Individual cells/mL to about 1.0x10 8 Individual cells/mL, e.g., at least or about 1.0x10 5 Individual cells/mL, 5X10 5 Individual cells/mL, 1X10 6 Individual cells/mL, 5X10 6 Individual cells/mL, 1X10 7 Individual cells/mL, 5X10 7 Individual cells/mL or 1x10 8 Individual cells/mL. In some embodiments, the composition of enriched T cells incubated with the stimulating agent comprises about 0.5x10 6 Individual cells/mL, 1X10 6 Individual cells/mL, 1.5x10 6 Individual cells/mL, 2X10 6 Individual cells/mL, 2.5x10 6 Individual cells/mL, 3X10 6 Individual cells/mL, 3.5x10 6 Individual cells/mL, 4x10 6 Individual cells/mL, 4.5x10 6 Individual cells/mL, 5X10 6 Individual cells/mL, 5.5x10 6 Individual cells/mL, 6X10 6 Individual cells/mL, 6.5x10 6 Individual cells/mL, 7X10 6 Individual cells/mL, 7.5x10 6 Individual cells/mL, 8x10 6 Individual cells/mL, 8.5x10 6 Individual cells/mL, 9X10 6 Individual cells/mL, 9.5x10 6 Individual cells/mL or 10x10 6 Individual cells/mL, e.g., about 2.4x10 6 Individual cells/mL.
In some embodiments, the T cell enriched composition is incubated with a stimulating agent at a temperature of from about 25 ℃ to about 38 ℃, such as from about 30 ℃ to about 37 ℃, for example, or about 37±2 ℃. In some embodiments, the enriched T cell composition is combined with the stimulating agent at from about 2.5% to about 7.5%, such as from about 4% to about 6%, e.g., at or about 5% ± 0.5% CO 2 Incubate together at the level. In some embodiments, the T cell enriched composition is contacted with a stimulating agent at a temperature of at or about 37℃and/or at or about 5% CO 2 Incubate together at the level.
In particular embodiments, the stimulating conditions comprise incubating, culturing and/or incubating the enriched T cell composition with and/or in the presence of one or more cytokines. In certain embodiments, the one or more cytokines are recombinant cytokines. In some embodiments, the one or more cytokines are human recombinant cytokines. In certain embodiments, one or more cytokines bind and/or are capable of binding to receptors expressed by and/or endogenous to T cells. In certain embodiments, the one or more cytokines are or include members of the 4-alpha-helix bundle family of cytokines. In some embodiments, members of the 4- α -helix bundle family of cytokines include, but are not limited to, interleukin 2 (IL-2), interleukin 4 (IL-4), interleukin 7 (IL-7), interleukin 9 (IL-9), interleukin 12 (IL-12), interleukin 15 (IL-15), granulocyte colony-stimulating factor (G-CSF), and granulocyte macrophage colony-stimulating factor (GM-CSF). In some embodiments, one or more cytokines is or includes IL-15. In certain embodiments, the one or more cytokines is or includes IL-7. In certain embodiments, the one or more cytokines is or includes IL-2. In some embodiments, the stimulation conditions include incubating the composition enriched in T cells (e.g., enriched in cd4+ T cells or enriched in cd8+ T cells) in the presence of a stimulating agent (anti-CD 3/anti-CD 28 magnetic beads) and in the presence of one or more recombinant cytokines as described.
In certain embodiments, the CD4+ T cell enriched composition is incubated with IL-2, e.g., recombinant IL-2. Without wishing to be bound by theory, particular embodiments contemplate that cd4+ T cells obtained from some subjects do not produce or do not sufficiently produce IL-2 in an amount that allows for growth, division, and expansion throughout the process of a composition for generating output cells (e.g., engineered cells suitable for use in cell therapy). In some embodiments, incubating the cd4+ T cell-enriched composition in the presence of recombinant IL-2 under stimulation increases the probability or likelihood that the cd4+ T cells of the composition will continue to survive, grow, expand, and/or activate during and throughout the incubation step. In some embodiments, incubating the enriched cd4+ T cell composition in the presence of recombinant IL-2 increases the probability and/or likelihood of producing an output composition of enriched cd4+ T cells (e.g., engineered cd4+ T cells suitable for cell therapy) from the enriched cd4+ T cell composition by at least 0.5%, at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least 7%, at least 8%, at least 9%, at least 10%, at least 11%, at least 12%, at least 13%, at least 14%, at least 15%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 100%, at least 150%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 10-fold, at least 25-fold, at least 50-fold, or at least 100-fold as compared to an alternative and/or exemplary method of incubating the enriched cd4+ T cell composition in the presence of recombinant IL-2.
In certain embodiments, the amount or concentration of one or more cytokines is measured and/or quantified in International Units (IU). The international units can be used to quantify vitamins, hormones, cytokines, vaccines, blood products and similar bioactive substances. In some embodiments, an IU is or includes a measure of biological agent efficacy by comparison to an international reference standard (e.g., WHO first international standard for human IL-2 (WHO 1st International Standard for Human IL-2), 86/504) having a particular weight and intensity. International units are the only accepted and standardized method of reporting bioactive units that are published and derived from international collaborative research work. In certain embodiments, the IU of the composition, sample, or source of cytokine may be obtained by a product comparison test with a similar WHO standard product. For example, in some embodiments, the IU/mg of a composition, sample or source of human recombinant IL-2, IL-7 or IL-15 is compared to the WHO standard IL-2 product (NIBSC code: 86/500), the WHO standard IL-17 product (NIBSC code: 90/530), and the WHO standard IL-15 product (NIBSC code: 95/554), respectively.
In some embodiments, the biological activity in IU/mg is equivalent to (ED in ng/ml 50 ) -1 x10 6 . In particular embodiments, recombinant human IL-2 or IL-15 ED 50 Equivalent to the concentration required for half maximal stimulation of cell proliferation (XTT cleavage) using CTLL-2 cells. In certain embodiments, the ED of recombinant human IL-7 50 Equivalent to the concentration required for half-maximal stimulation of PHA-activated human peripheral blood lymphocyte proliferation. Details relating to the determination and calculation of IU for IL-2 are discussed in Wadhwa et al, journal of Immunological Methods (2013), 379 (1-2): 1-7; and Gearing and Thorpe, journal of Immunological Methods (1988), 114 (1-2): 3-9; details relating to the determination and calculation of IU for IL-15 are discussed in Soman et al Journal of Immunological Methods (2009) 348 (1-2): 83-94; said document is hereby incorporated by reference in its entirety.
In certain embodiments, the compositions enriched for CD8+ T cells are incubated in the presence of IL-2 and/or IL-15 under stimulatory conditions. In certain embodiments, the composition enriched for CD4+ T cells is incubated in the presence of IL-2, IL-7 and/or IL-15 under stimulatory conditions. In some embodiments, IL-2, IL-7 and/or IL-15 is recombinant. In certain embodiments, IL-2, IL-7 and/or IL-15 is human. In particular embodiments, the one or more cytokines are or include human recombinant IL-2, IL-7, and/or IL-15. In some aspects, the incubation of the enriched T cell composition further comprises the presence of a stimulating agent, such as anti-CD 3/anti-CD 28 magnetic beads.
In some embodiments, the cells are incubated with a cytokine, such as a recombinant human cytokine, at a concentration of between 1IU/ml and 1,000IU/ml, between 10IU/ml and 50IU/ml, between 50IU/ml and 100IU/ml, between 100IU/ml and 200IU/ml, between 100IU/ml and 500IU/ml, between 250IU/ml and 500IU/ml, or between 500IU/ml and 1,000 IU/ml.
In some embodiments, the T cell enriched composition is incubated with IL-2, e.g., human recombinant IL-2, at a concentration of between 1IU/ml and 200IU/ml, between 10IU/ml and 100IU/ml, between 50IU/ml and 150IU/ml, between 80IU/ml and 120IU/ml, between 60IU/ml and 90IU/ml, or between 70IU/ml and 90 IU/ml. In particular embodiments, the T-cell enriched composition is incubated with recombinant IL-2 at a concentration of at or about 50IU/ml, 55IU/ml, 60IU/ml, 65IU/ml, 70IU/ml, 75IU/ml, 80IU/ml, 85IU/ml, 90IU/ml, 95IU/ml, 100IU/ml, 110IU/ml, 120IU/ml, 130IU/ml, 140IU/ml, or 150IU/ml. In some embodiments, the T cell enriched composition is incubated in the presence of recombinant IL-2 at or about 85 IU/ml. In some embodiments, the composition incubated with recombinant IL-2 is enriched for a population of T cells (e.g., cd4+ T cells and/or cd8+ T cells). In some embodiments, the T cell population is a cd4+ T cell population. In some embodiments, the T cell enriched composition is a cd8+ T cell enriched composition. In certain embodiments, the T cell enriched composition is enriched for cd8+ T cells, wherein the cd4+ T cells are not enriched and/or wherein the cd4+ T cells are negatively selected or depleted from the composition. In some embodiments, the T cell enriched composition is a cd4+ T cell enriched composition. In certain embodiments, the T cell enriched composition is enriched for cd4+ T cells, wherein the cd8+ T cells are not enriched and/or wherein the cd8+ T cells are negatively selected or depleted from the composition. In some embodiments, the enriched CD4+ T cell composition incubated with recombinant IL-2 may also be incubated with recombinant IL-7 and/or recombinant IL-15 as in the amounts described. In some embodiments, the enriched CD8+ T cell composition incubated with recombinant IL-2 may also be incubated with recombinant IL-15 as in the amounts described.
In some embodiments, the T cell enriched composition is incubated with recombinant IL-7, e.g., human recombinant IL-7, at a concentration of between 100IU/ml and 2,000IU/ml, between 500IU/ml and 1,000IU/ml, between 100IU/ml and 500IU/ml, between 500IU/ml and 750IU/ml, between 750IU/ml and 1,000IU/ml, or between 550IU/ml and 650 IU/ml. In particular embodiments, the T-cell enriched composition is incubated with recombinant IL-7 at a concentration of at or about 50IU/ml, 100IU/ml, 150IU/ml, 200IU/ml, 250IU/ml, 300IU/ml, 350IU/ml, 400IU/ml, 450IU/ml, 500IU/ml, 550IU/ml, 600IU/ml, 650IU/ml, 700IU/ml, 750IU/ml, 800IU/ml, 750IU/ml, or 1,000IU/ml. In a particular embodiment, the T cell enriched composition is incubated in the presence of recombinant IL-7 at or about 600 IU/ml. In some embodiments, the composition incubated with recombinant IL-7 is enriched for a population of T cells (e.g., cd4+ T cells). In some embodiments, the enriched CD4+ T cell composition incubated with recombinant IL-7 may also be incubated with recombinant IL-2 and/or recombinant IL-15 as in the amounts described. In certain embodiments, the T cell enriched composition is enriched for cd4+ T cells, wherein the cd8+ T cells are not enriched and/or wherein the cd8+ T cells are negatively selected or depleted from the composition. In some embodiments, the enriched CD8+ T cell composition is not incubated with recombinant IL-7.
In some embodiments, the T cell enriched composition is incubated with recombinant IL-15, e.g., human recombinant IL-15, at a concentration of between 0.1IU/ml and 100IU/ml, between 1IU/ml and 50IU/ml, between 5IU/ml and 25IU/ml, between 25IU/ml and 50IU/ml, between 5IU/ml and 15IU/ml, or between 10IU/ml and 100 IU/ml. In particular embodiments, the T-cell enriched composition is incubated with recombinant IL-15 at a concentration of at or about 1IU/ml, 2IU/ml, 3IU/ml, 4IU/ml, 5IU/ml, 6IU/ml, 7IU/ml, 8IU/ml, 9IU/ml, 10IU/ml, 11IU/ml, 12IU/ml, 13IU/ml, 14IU/ml, 15IU/ml, 20IU/ml, 25IU/ml, 30IU/ml, 40IU/ml, or 50IU/ml. In some embodiments, the T cell enriched composition is incubated in or at about 10IU/ml recombinant IL-15. In some embodiments, the composition incubated with recombinant IL-15 is enriched for a population of T cells (e.g., cd4+ T cells and/or cd8+ T cells). In some embodiments, the T cell population is a cd4+ T cell population. In some embodiments, the T cell enriched composition is a cd8+ T cell enriched composition. In certain embodiments, the T cell enriched composition is enriched for cd8+ T cells, wherein the cd4+ T cells are not enriched and/or wherein the cd4+ T cells are negatively selected or depleted from the composition. In some embodiments, the T cell enriched composition is a cd4+ T cell enriched composition. In certain embodiments, the T cell enriched composition is enriched for cd4+ T cells, wherein the cd8+ T cells are not enriched and/or wherein the cd8+ T cells are negatively selected or depleted from the composition. In some embodiments, the enriched CD4+ T cell composition incubated with recombinant IL-15 may also be incubated with recombinant IL-7 and/or recombinant IL-2 as in the amounts described. In some embodiments, the enriched CD8+ T cell composition incubated with recombinant IL-15 may also be incubated with recombinant IL-2 as in the amounts described.
In certain embodiments, cells (e.g., enriched cd4+ T cells and/or enriched cd8+ T cells) are incubated with a stimulating agent in the presence of one or more antioxidants. In some embodiments, antioxidants include, but are not limited to, one or more antioxidants including tocopherol, tocotrienol, alpha-tocopherol, beta-tocopherol, gamma-tocopherol, delta-tocopherol, alpha-tocotrienol, beta-tocotrienol, alpha-tocoquinone, trolox (6-hydroxy-2, 5,7, 8-tetramethylchroman-2-dicarboxylic acid), butylated Hydroxyanisole (BHA), butylated Hydroxytoluene (BHT), flavonoids, isoflavones, lycopene, beta-carotene, selenium, ubiquinone, syphilin (luetin), S-adenosylmethionine, glutathione, taurine, N-acetylcysteine (NAC), citric acid, L-carnitine, BHT, thioglycerol, ascorbic acid, propyl gallate, methionine, cysteine, homocysteine, glutathione, cystamine and cystathionine (cystathionine) and/or glycine-histidine. In some aspects, incubating the enriched T cell composition (e.g., enriched cd4+ T cells and/or enriched cd8+ T cells) with an antioxidant further comprises the presence of a stimulating agent (e.g., anti-CD 3/anti-CD 28 magnetic beads) and one or more recombinant cytokines (as described).
In some embodiments, the one or more antioxidants are or include sulfur-containing oxidizing agents. In certain embodiments, the sulfur-containing antioxidant may include a thiol-containing antioxidant and/or an antioxidant that exhibits one or more sulfur moieties, for example, within the ring structure. In some embodiments, the sulfur-containing antioxidants may include, for example, N-acetylcysteine (NAC) and 2, 3-Dimercaptopropanol (DMP), L-2-oxo-4-thiazolidine formate (OTC), and lipoic acid. In a particular embodiment, the sulfur-containing antioxidant is a glutathione precursor. In some embodiments, the glutathione precursor is a molecule that can be modified to a derivatized glutathione in one or more steps within the cell. In particular embodiments, glutathione precursors may include, but are not limited to, N-acetylcysteine (NAC), L-2-oxothiazolidine-4-carboxylic acid (procaysteine), lipoic acid, S-allylcysteine, or methioninesulfonium chloride.
In some embodiments, incubating the cells (e.g., enriched for cd4+ T cells and/or enriched for cd8+ T cells) under stimulation conditions comprises incubating the cells in the presence of one or more antioxidants. In certain embodiments, the cells are stimulated with a stimulating agent in the presence of one or more antioxidants. In some embodiments, cells are incubated in the presence of one or more antioxidants between 1ng/ml and 100ng/ml, between 10ng/ml and 1 μg/ml, between 100ng/ml and 10 μg/ml, between 1 μg/ml and 100 μg/ml, between 10 μg/ml and 1mg/ml, between 100 μg/ml and 1mg/ml, between 1 500 μg/ml and 2mg/ml, between 500 μg/ml and 5mg/ml, between 1mg/ml and 10mg/ml, or between 1mg/ml and 100 mg/ml. In some embodiments, cells are incubated in the presence of one or more antioxidants at or about 1ng/ml, 10ng/ml, 100ng/ml, 1 μg/ml, 10 μg/ml, 100 μg/ml, 0.2mg/ml, 0.4mg/ml, 0.6mg/ml, 0.8mg/ml, 1mg/ml, 2mg/ml, 3mg/ml, 4mg/ml, 5mg/ml, 10mg/ml, 20mg/ml, 25mg/ml, 50mg/ml, 100mg/ml, 200mg/ml, 300mg/ml, 400mg/ml, 500 mg/ml. In some embodiments, the one or more antioxidants are or include sulfur-containing antioxidants. In certain embodiments, the one or more antioxidants are or include glutathione precursors.
In some embodiments, the one or more antioxidants is or includes N-acetylcysteine (NAC). In some embodiments, incubating the cells (e.g., enriched for cd4+ T cells and/or enriched for cd8+ T cells) under stimulation conditions comprises incubating the cells in the presence of NAC. In certain embodiments, the cells are stimulated with a stimulating agent in the presence of NAC. In some embodiments, cells are incubated in the presence of NAC between 1ng/ml and 100ng/ml, between 10ng/ml and 1. Mu.g/ml, between 100ng/ml and 10. Mu.g/ml, between 1. Mu.g/ml and 100. Mu.g/ml, between 10. Mu.g/ml and 1mg/ml, between 100. Mu.g/ml and 1mg/ml, between 1-500. Mu.g/ml and 2mg/ml, between 500. Mu.g/ml and 5mg/ml, between 1mg/ml and 10mg/ml, or between 1mg/ml and 100 mg/ml. In some embodiments, cells are incubated in the presence of NAC at or about 1ng/ml, 10ng/ml, 100ng/ml, 1 μg/ml, 10 μg/ml, 100 μg/ml, 0.2mg/ml, 0.4mg/ml, 0.6mg/ml, 0.8mg/ml, 1mg/ml, 2mg/ml, 3mg/ml, 4mg/ml, 5mg/ml, 10mg/ml, 20mg/ml, 25mg/ml, 50mg/ml, 100mg/ml, 200mg/ml, 300mg/ml, 400mg/ml, 500 mg/ml. In some embodiments, the cells are incubated with or with about 0.8 mg/ml.
In certain embodiments, incubating the composition enriched for T cells (e.g., enriched for cd4+ T cells and/or enriched for cd8+ T cells) in the presence of one or more antioxidants, e.g., NAC, reduces activation in the cells as compared to cells incubated in an alternative and/or exemplary process in the absence of the antioxidants. In certain embodiments, reduced activation is measured by expression of one or more activation markers in the cell. In certain embodiments, activation markers include, but are not limited to, increased intracellular complexity (e.g., as determined by measuring Side Scatter (SSC)), increased cell size (e.g., as determined by measuring cell diameter and/or Forward Scatter (FSC)), increased CD27 expression, and/or decreased CD25 expression. In some embodiments, the cells of the composition have a negative, reduced, or low expression and/or extent of activation marker when examined during or after incubation, engineering, transduction, transfection, amplification, or formulation, or during or after any stage of the process that occurs after incubation. In some embodiments, the cells of the composition have a negative, reduced, or low expression and/or extent of activation markers after the process is complete. In certain embodiments, the cells outputting the composition have a negative, reduced, or low expression and/or extent of the activation marker.
In some embodiments, flow cytometry is used to determine the relative size of cells. In particular embodiments, FSC and SSC parameters are used to analyze cells and distinguish cells from one another based on size and internal complexity. In certain embodiments, particles or beads of known size may be measured as a standard for determining the actual size of the cells. In some embodiments, flow cytometry is used in combination with a staining agent, such as a labeled antibody, to measure or quantify expression of a surface protein (e.g., an activation marker, such as CD25 or CD 27).
In some embodiments, the composition enriched in T cells (e.g., enriched in cd4+ T cells and/or enriched in cd8+ T cells) is incubated in the presence of one or more antioxidants, e.g., NAC, and the cell diameter is reduced by at least 0.25 μm, 0.5 μm, 0.75 μm, 1.0 μm, 1.5 μm, 2 μm, 2.5 μm, 3 μm, 3.5 μm, 4 μm, 4.5 μm, 5 μm, or more than 5 μm as compared to cells incubated in an alternative and/or exemplary process in which incubation is not performed in the presence of the antioxidants. In certain embodiments, the composition enriched for T cells is incubated in the presence of one or more antioxidants, e.g., NAC, and the cell size is reduced by at least 1%, at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, or at least 90% as measured by FSC, as compared to cells incubated in an alternative and/or exemplary process in which incubation is not performed in the presence of the antioxidants.
In some embodiments, the composition enriched for T cells (e.g., enriched for cd4+ T cells and/or enriched for cd8+ T cells) is incubated in the presence of one or more antioxidants, e.g., NAC, and the intracellular complexity is reduced by at least 1%, at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, or at least 90% as measured by SSC, as compared to cells incubated in an alternative and/or exemplary process in which incubation is not performed in the presence of the antioxidants.
In particular embodiments, the composition enriched for T cells (e.g., enriched for cd4+ T cells and/or enriched for cd8+ T cells) is incubated in the presence of one or more antioxidants, e.g., NAC, and CD27 expression is reduced by at least 1%, at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% as measured, e.g., by flow cytometry, as compared to cells incubated in an alternative and/or exemplary procedure in which incubation is not performed in the presence of the antioxidants.
In certain embodiments, the composition enriched for T cells (e.g., enriched for cd4+ T cells and/or enriched for cd8+ T cells) is incubated in the presence of one or more antioxidants, e.g., NAC, and CD25 expression is increased by at least 1%, at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 100%, at least 150%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 10-fold, at least 25-fold, at least 50-fold, or at least 100-fold, e.g., as measured by flow cytometry, as compared to cells incubated in an alternative and/or exemplary procedure in which incubation is not performed in the presence of antioxidants.
In certain embodiments, incubating the composition enriched for T cells (e.g., enriched for cd4+ T cells and/or enriched for cd8+ T cells) in the presence of one or more antioxidants, e.g., NAC, increases expansion, e.g., during an incubation or incubation step or stage as described in section II-D. In some embodiments, the cell-enriched composition achieves 2-fold, 2.5-fold, 3-fold, 3.5-fold, 4-fold, 4.5-fold, 5-fold, 6-fold, 7-fold, 8-fold, 9-fold, 10-fold, or greater than 10-fold expansion within 14 days, 12 days, 10 days, 9 days, 8 days, 7 days, 6 days, 5 days, 4 days, or 3 days of initial incubation. In some embodiments, the cells of the composition undergo an expansion rate of at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 100%, at least 150%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 10-fold faster during incubation than incubated cells incubated in an alternative and/or exemplary procedure in which incubation is not performed in the presence of the antioxidant(s).
In certain embodiments, incubating the composition enriched for T cells (e.g., enriched for cd4+ T cells and/or enriched for cd8+ T cells) in the presence of one or more antioxidants, e.g., NAC, reduces the amount of cell death (e.g., due to apoptosis). In some embodiments, the composition enriched for T cells is incubated in the presence of one or more antioxidants, e.g., NAC, and at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or at least 99.9% of the cells survive, e.g., do not undergo apoptosis, for a period of 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or more than 7 days, or for at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or more than 7 days after the incubation is completed. In some embodiments, the composition is incubated in the presence of one or more antioxidants, e.g., NAC, and the cells of the composition have a survival rate that is at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 100%, at least 150%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 10-fold, at least 25-fold, at least 50-fold, or at least 100-fold greater than cells undergoing an exemplary and/or alternative process in which the cells are not incubated in the presence of the one or more antioxidants.
In certain embodiments, a composition enriched for T cells (e.g., enriched for cd4+ T cells and/or enriched for cd8+ T cells) is incubated in the presence of one or more antioxidants, e.g., NAC, and caspase expression, e.g., caspase 3 expression, is reduced by at least 1%, at least 5%, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% as compared to cells incubated in an alternative and/or exemplary process in which incubation is not performed in the presence of antioxidants.
In some embodiments, the composition or cell (e.g., enriched for cd4+ T cells and/or enriched for cd8+ T cells) is incubated in the presence of a stimulating condition or agent as described. Such conditions include those designed to induce proliferation, expansion, activation and/or survival of cells in a population to mimic antigen exposure and/or to elicit cells for genetic engineering (e.g., for the introduction of recombinant antigen receptors). Exemplary stimulating agents (e.g., anti-CD 3/anti-CD 28 magnetic beads) are described below. Incubation with the stimulating agent may also be performed in the presence of one or more stimulating cytokines, such as in the presence of one or more of recombinant IL-2, recombinant IL-7, and/or recombinant IL-15, and/or in the presence of at least one antioxidant, such as NAC, as described above. In some embodiments, the CD4+ T cell enriched composition is incubated with a stimulating agent (i.e., recombinant IL-2, recombinant IL-7, recombinant IL-15) and NAC in amounts as described under stimulating conditions. In some embodiments, the cd8+ T cell enriched composition is incubated with a stimulating agent (i.e., recombinant IL-2, recombinant IL-15) and NAC in amounts as described under stimulating conditions.
In some embodiments, the conditions for stimulation and/or activation may include one or more of the following: specific media, temperature, oxygen content, carbon dioxide content, time, agents (e.g., nutrients, amino acids, antibiotics, ions, and/or stimulatory factors (e.g., cytokines, chemokines, antigens, binding partners, fusion proteins, recombinant soluble receptors, and any other agents designed to activate cells)).
In some aspects, incubation is performed according to a variety of techniques, such as those described in the following documents: U.S. Pat. No. 6,040,1,77 to Riddell et al; klebaroff et al (2012) J Immunother.35 (9): 651-660; terakura et al (2012) blood.1:72-82; and/or Wang et al (2012) J Immunother35 (9): 689-701.
In some embodiments, at least a portion of the incubation performed in the presence of one or more stimulating conditions or agents is performed in the interior cavity of the centrifugal chamber, e.g., under centrifugal rotation, as described in international publication No. WO 2016/073602. In some embodiments, at least a portion of the incubation performed in the centrifugal chamber comprises mixing with one or more agents to induce stimulation and/or activation. In some embodiments, the cells (e.g., selected cells) are mixed with a stimulating condition or agent in the centrifugal chamber. In some aspects of such processes, a volume of cells is mixed with an amount of one or more stimulation conditions or agents that are much smaller than those typically used when performing similar stimulation in a cell culture plate or other system.
In some embodiments, the stimulus is added to the cells in the chamber cavity in an amount that is substantially smaller (e.g., no more than 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, or 80%) than the amount of stimulus that would normally be used or would be required to achieve about the same or similar efficiency of selection for the same cell number or same cell volume when selected, for example, in a tube or bag that is periodically oscillated or rotated, but not mixed in the centrifuge chamber. In some embodiments, incubation is performed with the addition of an incubation buffer to the cells and the stimulating agent to achieve a target volume of incubation of the reagent, e.g., about 10mL to about 200mL or about 20mL to about 125mL (e.g., at least or at least about or about 10mL, 20mL, 30mL, 40mL, 50mL, 60mL, 70mL, 80mL, 90mL, 100mL, 105mL, 110mL, 115mL, 120mL, 125mL, 130mL, 135mL, 140mL, 145mL, 150mL, 160mL, 170mL, 180mL, 190mL, or 200 mL). In some embodiments, the incubation buffer and the stimulating agent are pre-mixed prior to adding the cells. In some embodiments, the incubation buffer and the stimulating agent are added to the cells separately. In some embodiments, the stimulation incubation is performed under periodically mild mixing conditions, which may help promote energetically favorable interactions and thereby allow for less overall stimulator to be used while achieving stimulation and activation of the cells.
In some embodiments, incubation is typically performed under mixing conditions, such as in the presence of rotation, typically at a relatively low force or speed, such as a speed less than the speed used to precipitate the cells, such as from 600rpm to 1700rpm or from about 600rpm to about 1700rpm (e.g., at or about or at least 600rpm, 1000rpm, or 1500rpm, or 1700 rpm), such as at a RCF of from 80g to 100g or from about 80g to about 100g (e.g., at or about or at least 80g, 85g, 90g, 95g, or 100 g) of the sample or wall of the chamber or other container. In some embodiments, the rotation is performed using a repeating interval of such low speed rotation and a subsequent rest period, such as rotation and/or rest for 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 seconds, such as about 1 or 2 seconds, followed by rest for about 5, 6, 7, or 8 seconds.
In some embodiments, for example, the total duration of incubation with the stimulating agent is between or between about 1 hour and 96 hours, between 1 hour and 72 hours, between 1 hour and 48 hours, between 4 hours and 36 hours, between 8 hours and 30 hours, between 18 hours and 30 hours, or between 12 hours and 24 hours, such as at least or at least about or about 6 hours, 12 hours, 18 hours, 24 hours, 36 hours, or 72 hours. In some embodiments, further incubation is performed at or about between 1 hour and 48 hours, between 4 hours and 36 hours, between 8 hours and 30 hours, or between 12 hours and 24 hours, inclusive.
In some embodiments, the cells are cultured, incubated, and/or incubated under stimulating conditions prior to and/or during the step (as described by section II-C) for introducing the polynucleotide, e.g., a polynucleotide encoding a recombinant receptor, into the cells, e.g., by transduction and/or transfection. In certain embodiments, cells are cultured, incubated, and/or incubated under stimulating conditions for the following amounts of time prior to genetic engineering: between 30 minutes and 2 hours, between 1 hour and 8 hours, between 1 hour and 6 hours, between 6 hours and 12 hours, between 12 hours and 18 hours, between 16 hours and 24 hours, between 12 hours and 36 hours, between 24 hours and 48 hours, between 24 hours and 72 hours, between 42 hours and 54 hours, between 60 hours and 120 hours, between 96 hours and 120 hours, between 90 hours and between 1 day and 7 days, between 3 days and 8 days, between 1 day and 3 days, between 4 days and 6 days, or between 4 days and 5 days. In some embodiments, the cells are incubated for at or about 2 days prior to engineering.
In certain embodiments, the cells are incubated with and/or in the presence of a stimulating agent prior to and/or during genetically engineering the cells. In certain embodiments, the cells are incubated with and/or in the presence of a stimulating agent for the following amounts of time: between 12 hours and 36 hours, between 24 hours and 48 hours, between 24 hours and 72 hours, between 42 hours and 54 hours, between 60 hours and 120 hours, between 96 hours and 120 hours, between 90 hours and between 2 days and 7 days, between 3 days and 8 days, between 1 day and 8 days, between 4 days and 6 days or between 4 days and 5 days. In certain embodiments, the cells are cultured, incubated, and/or incubated under stimulating conditions for the following amounts of time prior to and/or during genetically engineering the cells: less than 10 days, 9 days, 8 days, 7 days, 6 days or 5 days, 4 days, or the amount of time as follows: less than 168 hours, 162 hours, 156 hours, 144 hours, 138 hours, 132 hours, 120 hours, 114 hours, 108 hours, 102 hours, or 96 hours. In particular embodiments, the cells are incubated with and/or in the presence of a stimulating agent for at least about 4 days, 5 days, 6 days, or 7 days. In some embodiments, the cells are incubated with and/or in the presence of a stimulating agent for at least about 4 days. In certain embodiments, the cells are incubated with and/or in the presence of a stimulating agent for at least about 5 days. In certain embodiments, the cells are incubated with and/or in the presence of a stimulating agent for less than 7 days.
In some embodiments, incubating the cells under stimulation conditions comprises incubating the cells with a stimulating agent as described in section II-B-1. In some embodiments, the stimulating agent comprises or includes beads, such as paramagnetic beads, and the cells are incubated with the stimulating agent at a ratio of less than 3:1 (beads: cells), such as a ratio of 1:1. In certain embodiments, the cells are incubated with the stimulating agent in the presence of one or more cytokines and/or one or more antioxidants. In some embodiments, the CD4+ T cell enriched composition is incubated with a stimulating agent in a ratio of 1:1 (beads: cells) in the presence of recombinant IL-2, IL-7, IL-15, and NAC. In certain embodiments, the CD8+ T cell enriched composition is incubated with a stimulating agent in a ratio of 1:1 (beads: cells) in the presence of recombinant IL-2, IL-15, and NAC. In some embodiments, the stimulating agent is removed and/or isolated from the cells within 6 days, 5 days, or 4 days, or within about 6 days, 5 days, or 4 days from the start or initiation of incubation, e.g., from the time the stimulating agent is added to or contacted with the cells.
1. Stimulating agent
In some embodiments, incubating the enriched cell composition under stimulation conditions is or includes incubating and/or contacting the enriched cell composition with a stimulating agent capable of activating and/or expanding T cells. In some embodiments, the stimulating agent is capable of stimulating and/or activating one or more signals in the cell. In some embodiments, one or more signals are mediated by a receptor. In particular embodiments, the one or more signals are, or are associated with, a change in the level or amount of signal transduction and/or second messengers (e.g., cAMP and/or intracellular calcium), a change in the amount, cellular localization, conformation, phosphorylation, ubiquitination, and/or truncation of one or more cellular proteins, and/or a change in cellular activity (e.g., transcription, translation, protein degradation, cellular morphology, activation state, and/or cell division). In particular embodiments, the stimulating agent activates and/or is capable of activating one or more intracellular signaling domains of one or more components of the TCR complex and/or one or more intracellular signaling domains of one or more co-stimulatory molecules.
In certain embodiments, the stimulating agent comprises particles (e.g., beads) conjugated or linked to one or more agents (e.g., biomolecules) capable of activating and/or expanding cells (e.g., T cells). In some embodiments, one or more agents are bound to the bead. In some embodiments, the beads are biocompatible, i.e., are composed of a material suitable for biological use. In some embodiments, the beads are non-toxic to cultured cells (e.g., cultured T cells). In some embodiments, the bead may be any particle capable of attaching an agent in a manner that allows for interaction between the agent and the cell.
In some embodiments, the stimulating agent contains one or more agents capable of activating and/or expanding cells (e.g., T cells) that are bound to or otherwise attached to the bead, such as to or attached to the surface of the bead. In certain embodiments, the beads are acellular particles. In particular embodiments, the beads may include colloidal particles, microspheres, nanoparticles, magnetic beads, and the like. In some embodiments, the beads are agarose beads. In certain embodiments, the beads are agarose gel beads.
In certain embodiments, the stimulating agent comprises monodisperse beads. In certain embodiments, the monodisperse beads comprise size dispersions having a standard deviation of diameter from each other of less than 5%.
In some embodiments, the beads contain one or more agents, such as agents coupled, conjugated or linked (directly or indirectly) to the surface of the beads. In some embodiments, an agent as contemplated herein may include, but is not limited to, RNA, DNA, a protein (e.g., an enzyme), an antigen, a polyclonal antibody, a monoclonal antibody, an antibody fragment, a carbohydrate, a lipid lectin, or any other biological molecule having affinity for a desired target. In some embodiments, the desired target is a T cell receptor and/or a component of a T cell receptor. In certain embodiments, the desired target is CD3. In certain embodiments, the desired target is a T cell costimulatory molecule, such as CD28, CD137 (4-1-BB), OX40, or ICOS. The one or more agents may be attached directly or indirectly to the bead by various methods known and available in the art. Attachment may be covalent, non-covalent, electrostatic or hydrophobic, and may be achieved by various attachment means including, for example, chemical means, mechanical means or enzymatic means. In some embodiments, a biomolecule (e.g., biotinylated anti-CD 3 antibody) may be indirectly attached to the bead through another biomolecule (e.g., an anti-biotin antibody) that is directly attached to the bead.
In some embodiments, the stimulating agent comprises a bead and one or more agents that interact directly with macromolecules on the cell surface. In certain embodiments, the beads (e.g., paramagnetic beads) interact with the cells through one or more agents (e.g., antibodies) that are specific for one or more macromolecules (e.g., one or more cell surface proteins) on the cells. In certain embodiments, the beads (e.g., paramagnetic beads) are labeled with a first agent (such as a primary antibody (e.g., an anti-biotin antibody) or other biomolecule) as described herein, followed by the addition of a second agent (such as a secondary antibody (e.g., a biotinylated anti-CD 3 antibody) or other second biomolecule (e.g., streptavidin), whereby the secondary antibody or other second biomolecule specifically binds to such primary antibody or other biomolecule on the particle.
In some embodiments, the stimulating agent contains one or more agents (e.g., antibodies) that are attached to the beads (e.g., paramagnetic beads) and that specifically bind to one or more of the following macromolecules on the cells (e.g., T cells): CD2, CD3, CD4, CD5, CD8, CD25, CD27, CD28, CD29, CD31, CD44, CD45RA, CD45RO, CD54 (ICAM-1), CD127, MHCI, MHCII, CTLA-4, ICOS, PD-1, OX40, CD27L (CD 70), 4-1BB (CD 137), 4-1BBL, CD30L, LIGHT, IL-2R, IL-12R, IL-1R, IL-15R; IFN-. Gamma. R, TNF-. Alpha. R, IL-4R, IL-10R, CD18/CDl la (LFA-1), CD62L (L-selectin), CD29/CD49d (VLA-4), notch ligands (e.g., delta-like 1/4, jagged 1/2, etc.), CCR1, CCR2, CCR3, CCR4, CCR5, CCR7, and CXCR3 or fragments thereof, including the corresponding ligands of these macromolecules or fragments thereof. In some embodiments, the agent (e.g., antibody) attached to the bead specifically binds to one or more of the following macromolecules on the cell (e.g., T cell): CD28, CD62L, CCR7, CD27, CD127, CD3, CD4, CD8, CD45RA and/or CD45RO. In some embodiments, the one or more agents attached to the bead are antibodies. Antibodies can include polyclonal antibodies, monoclonal antibodies (including full length antibodies with immunoglobulin Fc regions), antibody compositions with multi-epitope specificity, multi-specific antibodies (e.g., bispecific antibodies, diabodies, and single chain molecules), and antibody fragments (e.g., fab, F (ab') 2, and Fv). In some embodiments, the stimulating agent is an antibody fragment (including antigen binding fragments), such as a Fab, fab '-SH, fv, scFv, or (Fab') 2 fragment. It is to be understood that constant regions of any isotype can be used for antibodies contemplated herein, including IgG, igM, igA, igD and IgE constant regions, and that such constant regions can be obtained from any human or animal species (e.g., murine species).
In some embodiments, the agent is an antibody that binds to and/or recognizes one or more components of a T cell receptor. In certain embodiments, the agent is an anti-CD 3 antibody. In certain embodiments, the agent is an antibody that binds to and/or recognizes a co-receptor. In some embodiments, the stimulating agent comprises an anti-CD 28 antibody. In certain embodiments, the stimulatory agent comprises an anti-CD 3 antibody and an anti-CD 28 antibody. In some embodiments, the antibody is a Fab. In some embodiments, the stimulators contain an anti-CD 3 Fab and an anti-CD 28 Fab.
In some embodiments, the stimulators are anti-CD 3/anti-CD 28 streptavidin oligomer reagents, as described in PCT publication No. WO/2015/158868 or WO 2019/197949.
In some embodiments, the stimulating agent is anti-CD 3/anti-CD 28 beads (e.g.,m-450CD3/CD 28T cell expander and/or +.>Beads).
In some embodiments, the beads have a diameter of greater than about 0.001 μm, greater than about 0.01 μm, greater than about 0.1 μm, greater than about 1.0 μm, greater than about 10 μm, greater than about 50 μm, greater than about 100 μm, or greater than about 1000 μm and no more than about 1500 μm. In some embodiments, the beads have a diameter of about 1.0 μm to about 500 μm, about 1.0 μm to about 150 μm, about 1.0 μm to about 30 μm, about 1.0 μm to about 10 μm, about 1.0 μm to about 5.0 μm, about 2.0 μm to about 5.0 μm, or about 3.0 μm to about 5.0 μm. In some embodiments, the beads have a diameter of about 3 μm to about 5 μm. In some embodiments, the beads have a diameter of at least or at least about or about 0.001 μm, 0.01 μm, 0.1 μm, 0.5 μm, 1.0 μm, 1.5 μm, 2.0 μm, 2.5 μm, 3.0 μm, 3.5 μm, 4.0 μm, 4.5 μm, 5.0 μm, 5.5 μm, 6.0 μm, 6.5 μm, 7.0 μm, 7.5 μm, 8.0 μm, 8.5 μm, 9.0 μm, 9.5 μm, 10 μm, 12 μm, 14 μm, 16 μm, 18 μm, or 20 μm. In certain embodiments, the beads have a diameter of at or about 4.5 μm. In certain embodiments, the beads have a diameter of at or about 2.8 μm.
In some embodiments, the beads have a density greater than 0.001g/cm 3 Greater than 0.01g/cm 3 Greater than 0.05g/cm 3 Greater than 0.1g/cm 3 Greater than 0.5g/cm 3 Greater than 0.6g/cm 3 Greater than 0.7g/cm 3 Greater than 0.8g/cm 3 Greater than 0.9g/cm 3 More than 1g/cm 3 Greater than 1.1g/cm 3 Greater than 1.2g/cm 3 Greater than 1.3g/cm 3 Greater than 1.4g/cm 3 Greater than 1.5g/cm 3 More than 2g/cm 3 More than 3g/cm 3 Greater than 4g/cm 3 Or greater than 5g/cm 3 . In some embodiments, the density of the beads is about 0.001g/cm 3 And about 100g/cm 3 Between about 0.01g/cm 3 And about 50g/cm 3 Between about 0.1g/cm 3 And about 10g/cm 3 Between about 0.1g/cm 3 And about.5 g/cm 3 Between about 0.5g/cm 3 And about 1g/cm 3 Between about 0.5g/cm 3 And about 1.5g/cm 3 Between about 1g/cm 3 And about 1.5g/cm 3 Between about 1g/cm 3 And about 2g/cm 3 Between, or about 1g/cm 3 And about 5g/cm 3 Between them. In some embodiments, the beads have a density of about 0.5g/cm 3 About 0.5g/cm 3 About 0.6g/cm 3 About 0.7g/cm 3 About 0.8g/cm 3 About 0.9g/cm 3 About 1.0g/cm 3 About 1.1g/cm 3 About 1.2g/cm 3 About 1.3g/cm 3 About 1.4g/cm 3 About 1.5g/cm 3 About 1.6g/cm 3 About 1.7g/cm 3 About 1.8g/cm 3 About 1.9g/cm 3 Or about 2.0g/cm 3 . In certain embodiments, the beads have a density of about 1.6g/cm 3 . In particular embodiments, the beads or particles have a density of about 1.5g/cm 3 . In certain embodiments, the particles have a density of about 1.3g/cm 3
In certain embodiments, the plurality of beads have a uniform density. In certain embodiments, the uniform density comprises a standard deviation of density of less than 10%, less than 5%, or less than 1% of the average bead density.
In some embodiments, the surface area of the beads is about 0.001m 2 Particles/g (m) 2 /g) to about 1,000m 2 /g, about.010 m 2 /g to about 100m 2 /g, about 0.1m 2 /g to about 10m 2 /g, about 0.1m 2 /g to about 1m 2 /g, about 1m 2 /g to about 10m 2 /g, about 10m 2 /g to about 100m 2 /g, about 0.5m 2 /g to about 20m 2 /g, about 0.5m 2 /g to about 5m 2 /g or about 1m 2 /g to about 4m 2 Between/g. In some embodiments, the particles or beads have a surface area of about 1m 2 /g to about 4m 2 /g。
In some embodiments, the beads react in a magnetic field. In some embodiments, the beads are magnetic beads. In some embodiments, the magnetic beads are paramagnetic. In certain embodiments, the magnetic beads are superparamagnetic. In certain embodiments, the beads do not exhibit any magnetic properties unless they are exposed to a magnetic field.
In particular embodiments, the beads comprise a magnetic core, a paramagnetic core, or a superparamagnetic core. In some embodiments, the magnetic core comprises a metal. In some embodiments, the metal may be, but is not limited to, iron, nickel, copper, cobalt, gadolinium, manganese, tantalum, zinc, zirconium, or any combination thereof. In certain embodiments, the magnetic core comprises a metal oxide (e.g., iron oxide), ferrite (e.g., manganese ferrite, cobalt ferrite, nickel ferrite, etc.), hematite, and a metal alloy (e.g., coTaZn). In some embodiments, the magnetic core comprises one or more of ferrite, metal alloy, iron oxide, or chromium dioxide. In some embodiments, the magnetic core comprises elemental iron or a compound thereof. In some embodiments, the magnetic core comprises one or more of magnetite (Fe 3O 4), maghemite (γfe2o3), or pyrite (Fe 3S 4). In some embodiments, the core comprises iron oxide (e.g., fe 3 O 4 )。
In certain embodiments, the beads contain a magnetic core, paramagnetic core, and/or superparamagnetic core covered by a surface-functionalized coating (coat or coating). In some embodiments, the coating may contain the following materials: it may include, but is not limited to, polymers, polysaccharides, silica, fatty acids, proteins, carbon, agarose, sepharose, or combinations thereof. In some embodiments, the polymer may be polyethylene glycol, poly (lactic-co-glycolic acid), polyglutaridehyde, polyurethane, polystyrene, or polyvinyl alcohol. In certain embodiments, the outer coating (coat or coating) comprises polystyrene. In certain embodiments, the outer coating is surface functionalized.
In some embodiments, the stimulating agent comprises a bead comprising a metal oxide core (e.g., an iron oxide core) and a coating, wherein the metal oxide core comprises at least one polysaccharide (e.g., dextran), and wherein the coating comprises at least one polysaccharide (e.g., aminodextran), at least one polymer (e.g., polyurethane), and silica. In some embodiments, the metal oxide core is a colloidal iron oxide core. In certain embodiments, the one or more agents comprise an antibody or antigen-binding fragment thereof. In certain embodiments, the one or more agents include anti-CD 3 antibodies and anti-CD 28 antibodies or antigen-binding fragments thereof. In some embodiments, the stimulating agent comprises an anti-CD 3 antibody, an anti-CD 28 antibody, and an anti-biotin antibody. In some embodiments, the stimulating agent comprises an anti-biotin antibody. In some embodiments, the beads have a diameter of about 3 μm to about 10 μm. In some embodiments, the beads have a diameter of about 3 μm to about 5 μm. In certain embodiments, the beads have a diameter of about 3.5 μm.
In some embodiments, the stimulating agent comprises one or more agents attached to a bead comprising a metal oxide core (e.g., an iron oxide core) and a coating (e.g., a protective coating), wherein the coating comprises polystyrene. In certain embodiments, the beads are monodisperse paramagnetic (e.g., superparamagnetic) beads comprising paramagnetic (e.g., superparamagnetic) iron cores (e.g., comprising magnetite (Fe) 3 O 4 ) And/or maghemite (γfe) 2 O 3 ) A core) and a polystyrene coating (coat or coating). In some embodiments, the beads are non-porous. In some embodiments, the bead contains a functionalized surface to which one or more agents are attached. In certain embodiments, the one or more agents are covalently bound to the bead on the surface. In some embodiments, the one or more agents include an antibody or antigen-binding fragment thereof. In some embodiments, the one or more agents include anti-CD 3 antibodies and anti-CD 28 antibodies. In some embodiments, the stimulating agent is or comprises anti-CD 3/anti-CD 28 magnetic beads. In some embodiments, the one or more agents include an anti-CD 3 antibody and/or an anti-CD 28 antibody, and are capable ofAn antibody or fragment thereof that binds to a labeled antibody (e.g., a biotinylated antibody), such as a labeled anti-CD 3 or anti-CD 28 antibody. In certain embodiments, the beads have about 1.5g/cm 3 Density of about 1m 2 /g to about 4m 2 Surface area per gram. In particular embodiments; the beads are monodisperse superparamagnetic beads having a diameter of about 4.5 μm and a diameter of about 1.5g/cm 3 Is a density of (3). In some embodiments, the beads are of an average diameter of about 2.8 μm and about 1.3g/cm 3 Is a single dispersed superparamagnetic bead of a density of (1).
In some embodiments, the T cell enriched composition is incubated with the stimulating agent at or at a ratio of beads to cells of about 3:1, 2.5:1, 2:1, 1.5:1, 1.25:1, 1.2:1, 1.1:1, 1:1, 0.9:1, 0.8:1, 0.75:1, 0.67:1, 0.5:1, 0.3:1, or 0.2:1. In particular embodiments, the ratio of beads to cells is between 2.5:1 and 0.2:1, between 2:1 and 0.5:1, between 1.5:1 and 0.75:1, between 1.25:1 and 0.8:1, between 1.1:1 and 0.9:1. In particular embodiments, the ratio of stimulating agent to cells is about 1:1 or 1:1.
2. Removal of stimulating agents from cells
In certain embodiments, stimulating agents (e.g., anti-CD 3/anti-CD 28 magnetic beads) are removed and/or isolated from cells. Without wishing to be bound by theory, particular embodiments contemplate that in some cases, the binding and/or association between the stimulating agent and the cells may decrease over time during incubation. In certain embodiments, one or more agents may be added to reduce binding and/or association between the stimulating agent and the cells. In particular embodiments, a change in cell culture conditions (e.g., medium temperature or pH) can reduce binding and/or association between the stimulating agent and the cells. Thus, in some embodiments, the stimulating agent may be removed from the incubation, cell culture system, and/or solution separately from the cells, e.g., without removing the cells from the incubation, cell culture system, and/or solution as well.
Methods for removing stimulating agents (e.g., stimulating agents that are or contain particles such as beads or magnetizable particles) from cells are known. In some embodiments, the use of competing antibodies (e.g., unlabeled antibodies) that bind, for example, to a primary antibody of a stimulating agent and alter the affinity of the primary antibody for its antigen on a cell, thereby allowing for gentle desorption, may be used. In some cases, after desorption, the competing antibodies may remain associated with the particle (e.g., bead particle) while unreacted antibodies are washed off or may be washed off, and the cells are free of isolated, selected, enriched, and/or activated antibodies. An example of such an agent is DETACaBEAD (Friedl et al 1995; entschladen et al 1997). In some embodiments, the particle (e.g., bead particle) may be removed in the presence of a cleavable linker (e.g., DNA linker), thereby conjugating the particle-bound antibody to the linker (e.g., CELLection, dynal). In some cases, the linker region provides a cleavable site to remove particles (e.g., bead particles) from the cells after isolation, e.g., by addition of DNase or other release buffers. In some embodiments, other enzymatic methods may also be employed to release particles (e.g., bead particles) from cells. In some embodiments, the particles (e.g., bead particles or magnetizable particles) are biodegradable.
In some embodiments, the stimulating agent is magnetic, paramagnetic and/or superparamagnetic, and/or contains magnetic, paramagnetic and/or superparamagnetic beads, and the stimulating agent may be removed from the cells by exposing the cells to a magnetic field. Examples of suitable devices containing magnets for generating magnetic fields include DynaMag CTS (Thermo Fisher), magnetic separators (Takara) and easy Sep magnets (Stem Cell Technologies).
In certain embodiments, the stimulating agent is removed or isolated from the cells prior to completion of the provided methods, e.g., prior to harvesting, collecting, and/or formulating the engineered cells produced by the methods provided herein. In some embodiments, the stimulating agent is removed and/or isolated from the cells prior to engineering (e.g., transducing or transfecting) the cells. In certain embodiments, the stimulating agent is removed and/or isolated from the cells after the step of engineering the cells. In certain embodiments, the stimulating agent is removed prior to incubating the cells, e.g., prior to incubating the engineered, e.g., transfected or transduced, cells under conditions that promote proliferation and/or expansion.
In certain embodiments, the stimulating agent is isolated and/or removed from the cells after a certain amount of time. In a particular embodiment, the amount of time is the amount of time from the start of and/or initial incubation under the stimulation conditions. In certain embodiments, the initiation of incubation is considered to be at or about the time that the cells are contacted with the stimulating agent and/or the medium or solution containing the stimulating agent. In particular embodiments, the stimulating agent is removed or isolated from the cells within 10, 9, 8, 7, 6, 5, 4, 3, or 2 days or within about 10, 9, 8, 7, 6, 5, 4, 3, or 2 days after the initiation or initial incubation. In particular embodiments, the stimulating agent is removed and/or isolated from the cells at or about 9, 8, 7, 6, 5, 4, 3 or 2 days after the initiation or initial incubation. In certain embodiments, the stimulating agent is removed and/or isolated from the cells at or about 168 hours, 162 hours, 156 hours, 144 hours, 138 hours, 132 hours, 120 hours, 114 hours, 108 hours, 102 hours, or 96 hours after the initiation or initial incubation. In certain embodiments, the stimulating agent is removed and/or isolated from the cells at or about 5 days after the initiation and/or initial incubation. In some embodiments, the stimulating agent is removed and/or isolated from the cells at or about 4 days after the initiation and/or initial incubation.
C. Engineered cells
In some embodiments, the provided methods involve administering cells expressing a recombinant antigen receptor to a subject suffering from a disease or disorder. Various methods for introducing genetically engineered components, such as recombinant receptors (e.g., CARs or TCRs), are well known and can be used with the provided methods and compositions. Exemplary methods include those for transferring nucleic acids encoding a receptor, including by virus (e.g., retrovirus or lentivirus), transduction, transposon, and electroporation.
Cells expressing the receptor and administered by the provided methods include engineered cells. Genetic engineering generally involves introducing nucleic acids encoding recombinant or engineered components into a composition containing cells, such as by retroviral transduction, transfection or transformation.
In some embodiments, the methods provided herein are used in combination with one or more compositions for engineering enriched T cells. In certain embodiments, engineering is or includes introducing a polynucleotide, such as a recombinant polynucleotide encoding a recombinant protein. In particular embodiments, the recombinant protein is a recombinant receptor, such as any of the receptors described in section II. The introduction of a nucleic acid molecule encoding a recombinant protein (e.g., recombinant receptor) into a cell can be performed using any of a number of known vectors. Such vectors include viral and nonviral systems, including lentiviral and gamma retroviral systems, as well as transposon-based systems, such as PiggyBac or sleep Beauy-based gene transfer systems. Exemplary methods include those for transferring nucleic acids encoding a receptor, including by virus (e.g., retrovirus or lentivirus), transduction, transposon, and electroporation. In some embodiments, engineering produces one or more engineered compositions enriched for T cells.
In certain embodiments, one or more compositions enriched for T cells are engineered, e.g., transduced or transfected, prior to incubating the cells, e.g., under conditions that promote proliferation and/or expansion, as provided by the methods provided in section II-D. In certain embodiments, the one or more compositions enriched for T cells are engineered after the one or more compositions have been stimulated, activated, and/or incubated under stimulation conditions (as described in the methods provided in section II-B). In particular embodiments, one or more of the compositions is a stimulated composition. In certain embodiments, one or more of the stimulated compositions have been previously cryogenically frozen and stored and thawed prior to engineering.
In certain embodiments, the one or more compositions of stimulated T cells are or comprise two separate stimulated compositions of enriched T cells. In particular embodiments, two separate compositions of enriched T cells, e.g., two separate compositions of enriched T cells that have been selected, isolated, and/or enriched from the same biological sample, are separately engineered. In certain embodiments, the two separate compositions comprise a composition enriched for cd4+ T cells. In a particular embodiment, the two separate compositions comprise a composition enriched for cd8+ T cells. In some embodiments, the two separate compositions enriched for cd4+ T cells and enriched for cd8+ T cells are separately genetically engineered, such as after incubation under stimulation conditions as described above. In some embodiments, a single composition enriched for T cells is genetically engineered. In certain embodiments, the single composition is a composition enriched for cd4+ T cells. In some embodiments, the single composition is a composition enriched for cd4+ and cd8+ T cells that have been pooled from separate compositions prior to engineering.
In some embodiments, the composition of engineered, e.g., transduced or transfected enriched cd4+ T cells (e.g., stimulated cd4+ T cells) comprises at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or at or about 100% cd4+ T cells. In certain embodiments, the engineered composition enriched in cd4+ T cells (e.g., stimulated cd4+ T cells) comprises less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1% or less than 0.01% cd8+ T cells, and/or is free of cd8+ T cells, and/or is free or substantially free of cd8+ T cells.
In some embodiments, the composition of engineered, e.g., transduced or transfected enriched cd8+ T cells (e.g., stimulated cd8+ T cells) comprises at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or at or about 100% cd8+ T cells. In certain embodiments, the engineered composition enriched in cd8+ T cells (e.g., stimulated cd8+ T cells) comprises less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1% or less than 0.01% cd4+ T cells, and/or is free of cd4+ T cells, and/or is free or substantially free of cd4+ T cells.
In some embodiments, separate compositions enriched for cd4+ and cd8+ T cells are combined into a single composition and genetically engineered, e.g., transduced or transfected. In certain embodiments, the separate engineered compositions enriched for cd4+ and enriched for cd8+ T cells are combined into a single composition after genetic engineering has been performed and/or completed. In particular embodiments, separate compositions enriched for cd4+ and enriched for cd8+ T cells (e.g., separate compositions stimulated for cd4+ and cd8+ T cells) are separately engineered and separately processed for T cell incubation and/or expansion after genetic engineering has been performed and/or completed.
In some embodiments, the introduction of a polynucleotide, e.g., a recombinant polynucleotide encoding a recombinant protein, is performed by contacting an enriched cd4+ or cd8+ T cell (e.g., a stimulated cd4+ or cd8+ T cell) with a viral particle containing the polynucleotide. In some embodiments, contacting can be achieved by centrifugation, such as spin seeding (e.g., centrifugal seeding). In some embodiments, the composition containing the cells, virus particles, and agent may be spun, typically at a relatively low force or speed, such as a speed lower than the speed used to precipitate the cells, such as from 600rpm to 1700rpm or from about 600rpm to about 1700rpm (e.g., at or about or at least 600rpm, 1000rpm, or 1500rpm or 1700 rpm). In some embodiments, the rotation is performed with a force (e.g., relative centrifugal force) of from 100g to 3200g or from about 100g to about 3200g (e.g., or about or at least about 100g, 200g, 300g, 400g, 500g, 1000g, 1500g, 2000g, 2500g, 3000g, or 3200 g), such as or about 693g, as measured, for example, at an inner or outer wall of the chamber or cavity. The term "relative centrifugal force" or RCF is generally understood to be an effective force exerted on an object or substance (such as a cell, sample or pellet and/or a point in a rotated chamber or other container) relative to the gravitational force of the earth at a particular point in space as compared to a rotational axis. The values may be determined using well known formulas that take into account gravity, rotational speed and radius of rotation (distance from the axis of rotation and the object, substance or particle measuring the RCF). In some embodiments, at least a portion of the contacting, incubating, and/or engineering of the cells (e.g., cells from the enriched cd4+ T cells or the stimulated composition of enriched cd8+ T cells) with the virus is performed under rotation between about 100g and 3200g, 1000g and 2000g, 1000g and 3200g, 500g and 1000g, 400g and 1200g, 600g and 800g, 600g and 700g, or 500g and 700 g. In some embodiments, the rotation is between 600g and 700g, for example at or about 693 g.
In certain embodiments, at least a portion of the engineering, transduction, and/or transfection is performed under rotation, such as rotary seeding and/or centrifugation. In some embodiments, the rotation is performed, performed about, or performed for at least or at least about 5 minutes, 10 minutes, 15 minutes, 30 minutes, 60 minutes, 90 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 6 hours, 8 hours, 12 hours, 24 hours, 48 hours, 72 hours, 2 days, 3 days, 4 days, 5 days, 6 days, or performed for at least 7 days. In some embodiments, the rotation is performed for at least about 60 minutes. In certain embodiments, the rotation is performed for about 30 minutes. In some embodiments, the rotation is performed between 600g and 700g, for example at or about 693g, for about 30 minutes.
In certain embodiments, the number of living cells to be engineered, transduced and/or transfected ranges from about 5x10 6 Individual cells to about 100x10 7 Individual cells, e.g. from about 10x10 6 Individual cells to about 100x10 6 Individual cells, from about 100x10 6 From about 200x10 cells 6 Individual cells, from about 200x10 6 Individual cells to about 300x10 6 Individual cells, from about 300x10 6 Individual cells to about 400x10 6 Individual cells, from about 400x10 6 From about 500x10 per cell 6 Individual cells or from about 500x10 6 Individual cells to about 100x10 7 Individual cells. In particular examples, the number of living cells to be engineered, transduced and/or transfected is about or less than about 300x10 6 Individual cells.
In certain embodiments, at least a portion of the engineering, transduction, and/or transfection is performed at a volume (e.g., a rotary seeding volume) of from about 5mL to about 100mL, such as from about 10mL to about 50mL, from about 15mL to about 45mL, from about 20mL to about 40mL, from about 25mL to about 35mL, or at or about 30mL. In certain embodiments, the volume of the cell pellet after rotary seeding ranges from about 1mL to about 25mL, such as from about 5mL to about 20mL, from about 5mL to about 15mL, from about 5mL to about 10mL, or is about 10mL.
In some embodiments, gene transfer is accomplished by: cells are first stimulated, such as by combining them with a stimulus that induces a response (e.g., proliferation, survival, and/or activation), e.g., as measured by expression of a cytokine or activation marker, and then the activated cells are transduced and expanded in culture to an amount sufficient for clinical use. In certain embodiments, gene transfer is accomplished by: cells are first incubated under stimulating conditions, such as by any of the methods described in section I-B.
In some embodiments, the method for genetic engineering is performed by contacting one or more cells of the composition with a nucleic acid molecule encoding a recombinant protein (e.g., a recombinant receptor). In some embodiments, contacting can be achieved by centrifugation, such as rotary seeding (e.g., centrifugal seeding). Such methods include any of those described in International publication No. WO 2016/073602. Exemplary centrifugal chambers include those produced and sold by Biosafe SA, including forAnd->2, including a-200/F and a-200 centrifugal chambers, and various kits for use in such systems. Exemplary chambers, systems, and process instruments and cabinets are described in, for example, the following documents: U.S. patent No. 6,123,655, U.S. patent No. 6,733,433 and published U.S. patent application publication No. US2008/0171951, and published international patent application publication No. WO 00/38762, each of which are incorporated herein by reference in their entiretyThe content is incorporated by reference herein in its entirety. Exemplary kits for use with such systems include, but are not limited to, disposable kits sold under the product names CS-430.1, CS-490.1, CS-600.1, or CS-900.2 by BioSafe SA.
In some embodiments, the system is included with and/or placed in association with other instruments, including instruments for operating, automating, controlling and/or monitoring the transduction step and one or more various other processing steps performed in the system (e.g., one or more processing steps performed by a centrifugal chamber system as described herein or in international publication No. WO 2016/073602 may be used or combined). In some embodiments, such instruments are housed in cabinets. In some embodiments, the instrument includes a cabinet including a housing containing control circuitry, a centrifuge, a cover, a motor, a pump, sensors, a display, and a user interface. Exemplary devices are described in U.S. patent No. 6,123,655, U.S. patent No. 6,733,433, and U.S. patent No. 2008/0171951.
In some embodiments, the system includes a series of containers, such as bags, tubes, stopcocks, clamps, connectors, and centrifugal chambers. In some embodiments, the container (e.g., bag) comprises one or more containers (e.g., bags) containing the cells and viral vector particles to be transduced in the same container or in separate containers (e.g., as a bag or a separate bag). In some embodiments, the system further comprises one or more containers (e.g., bags) containing a culture medium, such as a diluent and/or wash solution, that is drawn into the chamber and/or other components during the process to dilute, resuspend, and/or wash the components and/or compositions. The vessel may be connected at one or more locations in the system, such as at locations corresponding to the input line, diluent line, wash line, waste line, and/or output line.
In some embodiments, the chamber is associated with a centrifuge that is capable of effecting rotation of the chamber, such as about its axis of rotation. Transduction of the bound cells and/or rotation may occur before, during and/or after incubation in one or more other processing steps. Thus, in some embodiments, one or more of the individual processing steps are performed under rotation (e.g., under a particular force). The chamber is typically rotatable vertically or substantially vertically such that the chamber is placed vertically during centrifugation, and the side walls and shaft are vertical or substantially vertical, and the one or more end walls are horizontal or substantially horizontal.
In some embodiments, the cell-containing composition and the viral vector particle-containing composition, and optionally air, may be combined or mixed prior to providing the composition to the cavity. In some embodiments, the cell-containing composition and the viral vector particle-containing composition, and optionally air, are provided separately in a cavity and combined and mixed therein. In some embodiments, the cell-containing composition, the viral vector particle-containing composition, and optionally air may be provided to the internal cavity in any order. In any of such embodiments, the composition comprising the cell and viral vector particles is an input composition once combined or mixed together, whether the input composition is combined and/or mixed inside or outside the centrifugal chamber, and/or whether the cell and viral vector particles are provided to the centrifugal chamber together or separately (e.g., simultaneously or sequentially).
In some embodiments, in the transduction method, uptake of a volume of gas (e.g., air) is performed prior to incubating the cells and viral vector particles (e.g., spinning). In some embodiments, uptake of a volume of gas (e.g., air) is performed during incubation (e.g., rotation) of the cells and viral vector particles in the transduction method.
In some embodiments, the liquid volume of the cells or viral vector particles comprising the transduction composition, and optionally the volume of air, may be a predetermined volume. The volume may be a volume programmed into the system and/or controlled by circuitry associated with the system.
In some embodiments, the intake of the transduction composition and optionally a gas (e.g., air) is controlled manually, semi-automatically, and/or automatically until a desired or predetermined volume has been ingested into the interior cavity of the chamber. In some embodiments, a sensor associated with the system may detect liquid and/or gas flowing into and out of the centrifugal chamber, such as by its color, flow rate, and/or density, and may communicate with associated circuitry to stop or continue ingestion as needed until such desired or predetermined volume ingestion has been achieved. In some aspects, a sensor that is programmed or only capable of detecting liquid in the system, rather than gas (e.g., air), may be enabled to allow gas (e.g., air) to pass into the system without stopping ingestion. In some such embodiments, when gas (e.g., air) ingestion is desired, an opaque tube may be placed in the line near the sensor. In some embodiments, the intake of a gas (e.g., air) may be controlled manually.
In aspects of the provided methods, the internal cavity of the centrifugal chamber is subjected to high-speed rotation. In some embodiments, the rotation is effected before, simultaneously with, after or intermittently with the intake of the liquid input composition and optionally air. In some embodiments, the rotation is achieved after ingestion of the liquid input composition and optionally air. In some embodiments, the rotation is by centrifugation of the centrifugal chamber by a relative centrifugal force of or about or at least about 800g, 1000g, 1100g, 1500, 1600g, 1800g, 2000g, 2200g, 2500g, 3000g, 3500g, or 4000g at the inner surface of the sidewall of the interior cavity and/or at the surface layer of the cells. In some embodiments, the rotation is by centrifugation with a force of greater than or about 1100g, such as greater than or about 1200g, greater than or about 1400g, greater than or about 1600g, greater than or about 1800g, greater than or about 2000g, greater than or about 2400g, greater than or about 2800g, greater than or about 3000g, or greater than or about 3200 g. In some embodiments, the rotation is by centrifugation at or about 1600g of force.
In some embodiments, the transduction method comprises spinning or centrifuging the transduction composition and optionally air in a centrifuge chamber for greater than or about 5 minutes, such as greater than or about 10 minutes, greater than or about 15 minutes, greater than or about 20 minutes, greater than or about 30 minutes, greater than or about 45 minutes, greater than or about 60 minutes, greater than or about 90 minutes, or greater than or about 120 minutes. In some embodiments, the transduction composition and optionally air are spun or centrifuged in the centrifuge chamber for greater than 5 minutes, but for no more than 60 minutes, no more than 45 minutes, no more than 30 minutes, or no more than 15 minutes. In certain embodiments, transduction comprises rotation or centrifugation for at least about 60 minutes.
In some embodiments, the transduction method comprises rotating or centrifuging the transduction composition and optionally air in a centrifugal chamber for the following time: between or about 10 minutes and 60 minutes, between 15 minutes and 45 minutes, between 30 minutes and 60 minutes, or between 45 minutes and 60 minutes, each comprising an end value, and the rotating or centrifuging is performed with a force at least or greater than or about 1000g, 1100g, 1200g, 1400g, 1500g, 1600g, 1800g, 2000g, 2200g, 2400g, 2800g, 3200g, or 3600g of the inner cavity sidewall inner surface and/or at the cell surface layer. In particular embodiments, the transduction method comprises rotating or centrifuging the transduction composition (e.g., cells and viral vector particles) at or about 1600g for or about 60 minutes.
In some embodiments, the gas (e.g., air) in the cavity of the chamber is vented from the chamber. In some embodiments, the gas (e.g., air) is vented to a container that is operatively connected to the centrifugal chamber as part of a closed system. In some embodiments, the container is a free or empty container. In some embodiments, air (e.g., gas) in the cavity of the chamber is exhausted through a filter that is operatively connected to the interior cavity of the chamber by a sterile tubing string. In some embodiments, the air is expelled using a manual, semi-automatic, or automatic process. In some embodiments, air is expelled from the cavity prior to, simultaneously with, intermittently with, or subsequent to delivering (expression) the output composition comprising the incubated cells and viral vector particles (e.g., cells that have begun to transduce or cells that have been transduced with viral vectors) from the cavity of the chamber.
In some embodiments, transduction and/or other incubation is performed as or as part of a continuous or semi-continuous process. In some embodiments, the continuous process involves continuous ingestion of the cells and viral vector particles, such as the transduction composition (either as a single pre-existing composition, or by continuous drawing into the same vessel (e.g., cavity), mixing portions thereof), and/or continuous delivery or evacuation of liquid from the vessel, and optionally evacuation of gas (e.g., air), during at least a portion of incubation (e.g., while centrifugation). In some embodiments, the continuous ingestion and continuous delivery are performed at least partially simultaneously. In some embodiments, continuous uptake occurs during part of the incubation, e.g., during part of the centrifugation, and continuous transport occurs during separate parts of the incubation. The two may be alternated. Thus, continuous ingestion and delivery while incubation is performed may allow for processing (e.g., transduction) of a sample of greater total volume.
In some embodiments, the incubation is part of a continuous process, the method comprising during at least a portion of the incubation, effecting continuous ingestion of the transduction composition into the cavity during rotation of the chamber and during a portion of the incubation, effecting continuous delivery of liquid from the cavity and optionally venting of gas (e.g., air) through the at least one opening during rotation of the chamber.
In some embodiments, semi-continuous incubation is performed by alternating between: the intake of the composition into the cavity, incubation, delivery of liquid from the cavity and optionally evacuation of gas (e.g., air) from the cavity, such as to an output container, is effected, followed by intake of a subsequent (e.g., second, third, etc.) composition containing more cells and other reagents (e.g., viral vector particles) for processing, and repetition of the process. For example, in some embodiments, the incubating is part of a semi-continuous process, the method comprising, prior to incubating, effecting uptake of the transduction composition into the cavity through the at least one opening, and, after incubating, effecting delivery of the fluid from the cavity; effecting uptake of another transduction composition comprising a cell and a viral vector particle into the internal cavity; and incubating the further transduction composition in the internal cavity under conditions whereby cells in the further transduction composition are transduced by the carrier. The process can continue in an iterative fashion for many additional rounds. In this regard, semi-continuous or continuous methods may allow for the production of even larger volumes and/or numbers of cells.
In some embodiments, a portion of the transduction incubation is performed in a centrifuge chamber, which is performed under conditions that include rotation or centrifugation.
In some embodiments, the method comprises incubating, wherein another portion of the incubation of the cells and viral vector particles occurs without rotation or centrifugation, typically after at least a portion of the incubation including rotation or centrifugation of the chamber. In certain embodiments, the incubation of the cells and viral vector particles is performed without rotation or centrifugation for at least 1 hour, 6 hours, 12 hours, 24 hours, 32 hours, 48 hours, 60 hours, 72 hours, 90 hours, 96 hours, 3 days, 4 days, 5 days, or more than 5 days. In certain embodiments, incubation is performed for at or about 72 hours.
In some such embodiments, further incubation is effected under conditions such that the viral vector integrates into the host genome of one or more cells. It is assessed or determined whether incubation has resulted in integration of the viral vector particles into the host genome, and therefore the conditions for further incubation are determined empirically to be within the level of the skilled person. In some embodiments, viral vector integration into the host genome can be assessed by measuring the expression level of a recombinant protein (e.g., a heterologous protein) encoded by a nucleic acid contained in the viral vector particle genome after incubation. The expression level of the recombinant molecule can be assessed using a variety of well known methods, for example in the case of cell surface proteins, such as by affinity-based methods (e.g., immunoaffinity-based methods), such as by flow cytometry. In some examples, expression is measured by detecting the transduction markers and/or reporter constructs. In some embodiments, the nucleic acid encoding the truncated surface protein is included in a vector and used as a marker for its expression and/or enhancement.
In some embodiments, the composition containing the cells, carrier (e.g., viral particles), and reagent may be spun, typically at a relatively low force or speed, such as a speed lower than the speed used to precipitate the cells, such as from 600rpm to 1700rpm or from about 600rpm to about 1700rpm (e.g., at or about or at least 600rpm, 1000rpm, or 1500rpm or 1700 rpm). In some embodiments, the rotation is performed with a force (e.g., relative centrifugal force) of from 100g to 3200g or from about 100g to about 3200g (e.g., or about or at least about 100g, 200g, 300g, 400g, 500g, 1000g, 1500g, 2000g, 2500g, 3000g, or 3200 g), as measured, for example, at an inner or outer wall of the chamber or cavity. The term "relative centrifugal force" or RCF is generally understood to be an effective force exerted on an object or substance (such as a cell, sample or pellet and/or a point in a rotated chamber or other container) relative to the gravitational force of the earth at a particular point in space as compared to a rotational axis. The values may be determined using well known formulas that take into account gravity, rotational speed and radius of rotation (distance from the axis of rotation and the object, substance or particle measuring the RCF).
In some embodiments, the cells are transferred to a bioreactor bag assembly during at least a portion of the genetic engineering (e.g., transduction), and/or after genetic engineering, for culturing the genetically engineered cells, such as for culturing or expanding the cells, as described above.
In certain embodiments, the composition enriched for T cells is engineered, e.g., transduced or transfected, in the presence of a transduction adjuvant. In some embodiments, the composition of enriched T cells is engineered in the presence of one or more polycations. In some embodiments, transduction is performed in the presence of one or more transduction adjuvants, e.g., incubating the T cell enriched composition with viral vector particles. In certain embodiments, the T cell enriched composition is transfected in the presence of one or more transduction adjuvants, e.g., incubated with a non-viral vector. In certain embodiments, the presence of one or more transduction adjuvants increases the efficiency of gene delivery, such as by increasing the amount, fraction, and/or percentage of engineered (e.g., transduced or transfected) cells in the composition. In certain embodiments, the presence of one or more transduction adjuvants increases the efficiency of transfection. In certain embodiments, the presence of one or more transduction adjuvants increases the efficiency of transduction. In particular embodiments, at least 25%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% of the cells engineered in the presence of the polycation contain or express the recombinant polynucleotide. In some embodiments, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 100%, at least 150%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 10-fold, at least 25-fold, at least 50-fold, or at least 100-fold more cells in the composition are engineered to contain or express the recombinant transduction adjuvant than alternative and/or exemplary methods of engineering cells in the absence of the transduction adjuvant.
In some embodiments, the composition of enriched cells is engineered in the presence of less than 100 μg/ml, less than 90 μg/ml, less than 80 μg/ml, less than 75 μg/ml, less than 70 μg/ml, less than 60 μg/ml, less than 50 μg/ml, less than 40 μg/ml, less than 30 μg/ml, less than 25 μg/ml, less than 20 μg/ml, or less than μg/ml, less than 10 μg/ml of a transduction adjuvant. In certain embodiments, transduction adjuvants suitable for use in the provided methods include, but are not limited to, polycations, fibronectin or fragments or variants derived from fibronectin, retroNectin, and combinations thereof.
In some embodiments, the cells are engineered in the presence of cytokines, such as recombinant human cytokines, at a concentration of between 1IU/ml and 1,000IU/ml, between 10IU/ml and 50IU/ml, between 50IU/ml and 100IU/ml, between 100IU/ml and 200IU/ml, between 100IU/ml and 500IU/ml, between 250IU/ml and 500IU/ml, or between 500IU/ml and 1,000 IU/ml.
In some embodiments, the composition of enriched T cells is engineered in the presence of IL-2, e.g., human recombinant IL-2, at a concentration of between 1IU/ml and 200IU/ml, between 10IU/ml and 100IU/ml, between 50IU/ml and 150IU/ml, between 80IU/ml and 120IU/ml, between 60IU/ml and 90IU/ml, or between 70IU/ml and 90 IU/ml. In particular embodiments, the composition of enriched T cells is engineered in the presence of recombinant IL-2 at a concentration of at or about 50IU/ml, 55IU/ml, 60IU/ml, 65IU/ml, 70IU/ml, 75IU/ml, 80IU/ml, 85IU/ml, 90IU/ml, 95IU/ml, 100IU/ml, 110IU/ml, 120IU/ml, 130IU/ml, 140IU/ml, or 150IU/ml. In some embodiments, the enriched T cell composition is engineered in the presence of at or about 85 IU/ml. In some embodiments, the T cell population is a cd4+ T cell population. In certain embodiments, the T cell enriched composition is enriched for cd4+ T cells, wherein the cd8+ T cells are not enriched and/or wherein the cd8+ T cells are negatively selected or depleted from the composition. In a particular embodiment, the T cell enriched composition is a cd8+ T cell enriched composition. In certain embodiments, the T cell enriched composition is enriched for cd8+ T cells, wherein the cd4+ T cells are not enriched and/or wherein the cd4+ T cells are negatively selected or depleted from the composition.
In some embodiments, the composition of enriched T cells is engineered in the presence of recombinant IL-7, e.g., human recombinant IL-7, at a concentration of between 100IU/ml and 2,000IU/ml, between 500IU/ml and 1,000IU/ml, between 100IU/ml and 500IU/ml, between 500IU/ml and 750IU/ml, between 750IU/ml and 1,000IU/ml, or between 550IU/ml and 650 IU/ml. In particular embodiments, the composition of enriched T cells is engineered in the presence of IL-7 at a concentration of at or about 50IU/ml, 100IU/ml, 150IU/ml, 200IU/ml, 250IU/ml, 300IU/ml, 350IU/ml, 400IU/ml, 450IU/ml, 500IU/ml, 550IU/ml, 600IU/ml, 650IU/ml, 700IU/ml, 750IU/ml, 800IU/ml, 750IU/ml, or 1,000IU/ml. In a particular embodiment, the enriched T cell composition is engineered in the presence of IL-7 at or about 600 IU/ml. In some embodiments, a composition engineered in the presence of recombinant IL-7 is enriched for a population of T cells (e.g., cd4+ T cells). In certain embodiments, the T cell enriched composition is enriched for cd4+ T cells, wherein the cd8+ T cells are not enriched and/or wherein the cd8+ T cells are negatively selected or depleted from the composition.
In some embodiments, the composition of enriched T cells is engineered in the presence of recombinant IL-15, e.g., human recombinant IL-15, at a concentration of between 0.1IU/ml and 100IU/ml, between 1IU/ml and 50IU/ml, between 5IU/ml and 25IU/ml, between 25IU/ml and 50IU/ml, between 5IU/ml and 15IU/ml, or between 10IU/ml and 100 IU/ml. In particular embodiments, the composition of enriched T cells is engineered in the presence of IL-15 at a concentration of at or about 1IU/ml, 2IU/ml, 3IU/ml, 4IU/ml, 5IU/ml, 6IU/ml, 7IU/ml, 8IU/ml, 9IU/ml, 10IU/ml, 11IU/ml, 12IU/ml, 13IU/ml, 14IU/ml, 15IU/ml, 20IU/ml, 25IU/ml, 30IU/ml, 40IU/ml, or 50IU/ml. In some embodiments, the T cell enriched composition is engineered in or in about 10IU/ml IL-15. In some embodiments, the T cell enriched composition is incubated in or at about 10IU/ml recombinant IL-15. In some embodiments, a composition engineered in the presence of recombinant IL-15 is enriched for a population of T cells (e.g., cd4+ T cells and/or cd8+ T cells). In some embodiments, the T cell enriched composition is a cd8+ T cell enriched composition. In certain embodiments, the T cell enriched composition is enriched for cd8+ T cells, wherein the cd4+ T cells are not enriched and/or wherein the cd4+ T cells are negatively selected or depleted from the composition. In some embodiments, the T cell enriched composition is a cd4+ T cell enriched composition. In certain embodiments, the T cell enriched composition is enriched for cd4+ T cells, wherein the cd8+ T cells are not enriched and/or wherein the cd8+ T cells are negatively selected or depleted from the composition.
In certain embodiments, compositions enriched for CD8+ T cells are engineered in the presence of IL-2 and/or IL-15. In certain embodiments, the composition enriched for CD4+ T cells is engineered in the presence of IL-2, IL-7, and/or IL-15. In some embodiments, IL-2, IL-7 and/or IL-15 is recombinant. In certain embodiments, IL-2, IL-7 and/or IL-15 is human. In particular embodiments, the one or more cytokines are or include human recombinant IL-2, IL-7, and/or IL-15.
In certain embodiments, the cells are engineered in the presence of one or more antioxidants. In some embodiments, antioxidants include, but are not limited to, one or more antioxidants including tocopherol, tocotrienol, alpha-tocopherol, beta-tocopherol, gamma-tocopherol, delta-tocopherol, alpha-tocotrienol, beta-tocotrienol, alpha-tocoquinone, trolox (6-hydroxy-2, 5,7, 8-tetramethylchroman-2-dicarboxylic acid), butylated Hydroxyanisole (BHA), butylated Hydroxytoluene (BHT), flavonoids, isoflavones, lycopene, beta-carotene, selenium, ubiquinone, syphilin, S-adenosylmethionine, glutathione, taurine, N-acetylcysteine (NAC), citric acid, L-carnitine, BHT, thioglycerol, ascorbic acid, propyl gallate, methionine, cysteine, homocysteine, glutathione, cystamine, and cystathionine, and/or glycine-histidine.
In some embodiments, the one or more antioxidants are or include sulfur-containing oxidizing agents. In certain embodiments, the sulfur-containing antioxidant may include a thiol-containing antioxidant and/or an antioxidant that exhibits one or more sulfur moieties, for example, within the ring structure. In some embodiments, the sulfur-containing antioxidants may include, for example, N-acetylcysteine (NAC) and 2, 3-Dimercaptopropanol (DMP), L-2-oxo-4-thiazolidine formate (OTC), and lipoic acid. In a particular embodiment, the sulfur-containing antioxidant is a glutathione precursor. In some embodiments, the glutathione precursor is a molecule that can be modified to a derivatized glutathione in one or more steps within the cell. In particular embodiments, glutathione precursors may include, but are not limited to, N-acetylcysteine (NAC), L-2-oxothiazolidine-4-carboxylic acid (procaysteine), lipoic acid, S-allylcysteine, or methioninesulfonium chloride.
In some embodiments, the cells are engineered in the presence of one or more antioxidants. In some embodiments, the cells are engineered in the presence of one or more antioxidants between 1ng/ml and 100ng/ml, between 10ng/ml and 1 μg/ml, between 100ng/ml and 10 μg/ml, between 1 μg/ml and 100 μg/ml, between 10 μg/ml and 1mg/ml, between 100 μg/ml and 1mg/ml, between 500 μg/ml and 2mg/ml, between 500 μg/ml and 5mg/ml, between 1mg/ml and 10mg/ml, or between 1mg/ml and 100 mg/ml. In some embodiments, the cells are engineered in the presence of one or more antioxidants at or about 1ng/ml, 10ng/ml, 100ng/ml, 1 μg/ml, 10 μg/ml, 100 μg/ml, 0.2mg/ml, 0.4mg/ml, 0.6mg/ml, 0.8mg/ml, 1mg/ml, 2mg/ml, 3mg/ml, 4mg/ml, 5mg/ml, 10mg/ml, 20mg/ml, 25mg/ml, 50mg/ml, 100mg/ml, 200mg/ml, 300mg/ml, 400mg/ml, 500 mg/ml. In some embodiments, the one or more antioxidants are or include sulfur-containing antioxidants. In certain embodiments, the one or more antioxidants are or include glutathione precursors.
In some embodiments, the cells are engineered in the presence of NAC. In some embodiments, cells are engineered in the presence of NAC between 1ng/ml and 100ng/ml, between 10ng/ml and 1 μg/ml, between 100ng/ml and 10 μg/ml, between 1 μg/ml and 100 μg/ml, between 10 μg/ml and 1mg/ml, between 100 μg/ml and 1mg/ml, between 1,500 μg/ml and 2mg/ml, between 500 μg/ml and 5mg/ml, between 1mg/ml and 10mg/ml, or between 1mg/ml and 100 mg/ml. In some embodiments, the cells are engineered in the presence of NAC at or about 1ng/ml, 10ng/ml, 100ng/ml, 1 μg/ml, 10 μg/ml, 100 μg/ml, 0.2mg/ml, 0.4mg/ml, 0.6mg/ml, 0.8mg/ml, 1mg/ml, 2mg/ml, 3mg/ml, 4mg/ml, 5mg/ml, 10mg/ml, 20mg/ml, 25mg/ml, 50mg/ml, 100mg/ml, 200mg/ml, 300mg/ml, 400mg/ml, 500 mg/ml. In some embodiments, the cells are engineered with or with about 0.8 mg/ml.
In some embodiments, the composition of enriched T cells (e.g., stimulated T cells, e.g., stimulated cd4+ T cells or stimulated cd8+ T cells) is engineered in the presence of one or more polycations. In some embodiments, the composition enriched for T cells (e.g., stimulated T cells, e.g., stimulated cd4+ T cells or stimulated cd8+ T cells) is transduced, e.g., incubated with a viral vector particle, in the presence of one or more polycations. In certain embodiments, a composition of enriched T cells (e.g., stimulated T cells, e.g., stimulated cd4+ T cells or stimulated cd8+ T cells) is transfected with a non-viral vector (e.g., incubated with a non-viral vector) in the presence of one or more polycations. In certain embodiments, the presence of one or more polycations increases the efficiency of gene delivery, such as by increasing the amount, fraction, and/or percentage of engineered (e.g., transduced or transfected) cells in the composition. In certain embodiments, the presence of one or more polycations increases the efficiency of transfection. In certain embodiments, the presence of one or more polycations increases the efficiency of transduction. In particular embodiments, at least 25%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% of the cells engineered in the presence of the polycation contain or express the recombinant polynucleotide. In some embodiments, at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 100%, at least 150%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 10-fold, at least 25-fold, at least 50-fold, or at least 100-fold more cells in the composition are engineered to contain or express the recombinant polynucleotide than alternative and/or exemplary methods of engineering cells in the absence of the polycation.
In certain embodiments, for example, the cell-enriched composition, e.g., the cd4+ T cell-enriched or cd8+ T cell-enriched (e.g., stimulated T cell thereof) composition, is engineered in the presence of a low concentration or amount of polycation relative to an exemplary and/or alternative method of engineering cells in the presence of a polyanion. In certain embodiments, enriched cells, such as compositions of stimulated T cells (e.g., stimulated cd4+ T cells or stimulated cd8+ T cells), are engineered in the presence of an amount or concentration of less than 90%, less than 80%, less than 75%, less than 70%, less than 60%, less than 50%, less than 40%, less than 30%, less than 25%, less than 20%, less than 10%, less than 5%, less than 1%, less than 0.1%, or less than 0.01% of the polycation used in the exemplary and/or alternative process of engineering cells. In some embodiments, the composition of enriched cells, such as stimulated T cells (e.g., stimulated cd4+ T cells or stimulated cd8+ T cells), is engineered in the presence of less than 100 μg/ml, less than 90 μg/ml, less than 80 μg/ml, less than 75 μg/ml, less than 70 μg/ml, less than 60 μg/ml, less than 50 μg/ml, less than 40 μg/ml, less than 30 μg/ml, less than 25 μg/ml, less than 20 μg/ml, or less than μg/ml, less than 10 μg/ml of polycation. In particular embodiments, the composition of enriched cells (e.g., stimulated T cells, e.g., stimulated CD4+ T cells or stimulated CD8+ T cells) is engineered in the presence of polycations at or about 1 μg/ml, 5 μg/ml, 10 μg/ml, 15 μg/ml, 20 μg/ml, 25 μg/ml, 30 μg/ml, 35 μg/ml, 40 μg/ml, 45 μg/ml, or 50 μg/ml.
In certain embodiments, engineering a composition of enriched cells, such as stimulated T cells (e.g., stimulated cd4+ T cells or stimulated cd8+ T cells), in the presence of a polycation reduces the amount of cell death (e.g., due to necrosis, apoptosis, or apoptosis). In some embodiments, a composition of enriched T cells, such as stimulated T cells (e.g., stimulated cd4+ T cells or stimulated cd8+ T cells), is engineered in the presence of a low amount of polycation (e.g., less than 100 μg/ml, 50 μg/ml, or 10 μg/ml), and 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or more than 7 days, or at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or more than 7 days, at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or at least 99.9% of the cells survive, e.g., do not undergo necrosis, programmed cell death, or apoptosis, after the engineering step is completed. In some embodiments, the composition is engineered in the presence of a low concentration or amount of polycation compared to alternative and/or exemplary methods of engineering cells in the presence of a higher amount or concentration of polycation (e.g., greater than 50 μg/ml, 100 μg/ml, 500 μg/ml, or 1,000 μg/ml), and the cells of the composition have a viability that is at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 100%, at least 150%, at least 1 fold, at least 2 fold, at least 3 fold, at least 4 fold, at least 5 fold, at least 10 fold, at least 25 fold, at least 50 fold, or at least 100 fold greater than cells undergoing the exemplary and/or alternative processes.
In some embodiments, the polycation is positively charged. In certain embodiments, the polycation reduces the repulsive force between the cell and the vector (e.g., viral or non-viral vector) and mediates contact and/or binding of the vector to the cell surface. In some embodiments, the polycation is a polybrene, DEAE-dextran, protamine sulfate, poly-L-lysine, or cationic liposome.
In a particular embodiment, the polycation is protamine sulfate. In some embodiments, the composition of enriched T cells, such as stimulated T cells (e.g., cd4+ T cells stimulated or cd8+ T cells stimulated) is engineered in the presence of less than or about 500 μg/ml, less than or about 400 μg/ml, less than or about 300 μg/ml, less than or about 200 μg/ml, less than or about 150 μg/ml, less than or about 100 μg/ml, less than or about 90 μg/ml, less than or about 80 μg/ml, less than or about 75 μg/ml, less than or about 70 μg/ml, less than or about 60 μg/ml, less than or about 50 μg/ml, less than or about 40 μg/ml, less than or about 30 μg/ml, less than or about 25 μg/ml, less than or about 20 μg/ml, or less than or about 15 μg/ml, or less than or about 10 μg/ml of protamine sulfate. In particular embodiments, the composition of enriched cells, such as stimulated T cells (e.g., stimulated CD4+ T cells or stimulated CD8+ T cells), is engineered in the presence of sulfuric acid protamine at or about 1 μg/ml, 5 μg/ml, 10 μg/ml, 15 μg/ml, 20 μg/ml, 25 μg/ml, 30 μg/ml, 35 μg/ml, 40 μg/ml, 45 μg/ml, 50 μg/ml, 55 μg/ml, 60 μg/ml, 75 μg/ml, 80 μg/ml, 85 μg/ml, 90 μg/ml, 95 μg/ml, 100 μg/ml, 105 μg/ml, 110 μg/ml, 115 μg/ml, 120 μg/ml, 125 μg/ml, 130 μg/ml, 135 μg/ml, 140 μg/ml, 145 μg/ml, or 150 μg/ml.
In some embodiments, an engineered composition enriched for cd4+ T cells, such as stimulated T cells (e.g., stimulated cd4+ T cells), comprises at least 40%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or at or about 100% cd4+ T cells. In certain embodiments, the composition of engineered enriched cd4+ T cells, such as stimulated T cells (e.g., stimulated cd4+ T cells), comprises less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1%, or less than 0.01% cd8+ T cells, and/or is free of cd8+ T cells, and/or is free or substantially free of cd8+ T cells.
In some embodiments, the composition of engineered enriched cd8+ T cells, such as stimulated T cells (e.g., stimulated cd8+ T cells), comprises at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or at or about 100% cd8+ T cells. In certain embodiments, the composition of engineered enriched cd8+ T cells, such as stimulated T cells (e.g., stimulated cd8+ T cells), comprises less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1%, or less than 0.01% cd4+ T cells, and/or is free of cd4+ T cells, and/or is free or substantially free of cd4+ T cells.
In some embodiments, engineering the cells includes culturing, contacting, or incubating with a vector (e.g., a viral vector or a non-viral vector). In certain embodiments, engineering comprises culturing, contacting and/or incubating the cells with the carrier, performing, or performing for about or at least 4 hours, 6 hours, 8 hours, 12 hours, 16 hours, 18 hours, 24 hours, 30 hours, 36 hours, 40 hours, 48 hours, 54 hours, 60 hours, 72 hours, 84 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, or 7 days or more than 7 days. In particular embodiments, engineering comprises culturing, contacting and/or incubating the cells with the carrier for at or about 24 hours, 36 hours, 48 hours, 60 hours, 72 hours, or 84 hours, or for at or about 2 days, 3 days, 4 days, or 5 days. In some embodiments, the engineering step is performed for at least about 24 hours, 36 hours, 48 hours, 60 hours, 72 hours, or 84 hours. In certain embodiments, the engineering is performed for about 60 hours or about 84 hours, for about 72 hours, or for about 2 days.
In some embodiments, the engineering is performed at a temperature of from about 25 ℃ to about 38 ℃, such as from about 30 ℃ to about 37 ℃, from about 36 ℃ to about 38 ℃, or at or about 37±2 ℃. In some embodiments, the composition enriched in T cells is at a CO of from about 2.5% to about 7.5%, such as from about 4% to about 6%, for example, at or about 5% ± 0.5% 2 Engineering at the level. In some embodiments, the T cell enriched composition is at or about 37℃and/or at or about 5% CO 2 Engineering at the level.
In some embodiments, the cells are incubated after one or more steps for genetically engineering, e.g., transducing or transfecting, the cells (e.g., cd4+ and/or cd8+ T cells) to contain the polynucleotide encoding the recombinant receptor. In some embodiments, incubating may include culturing, incubating, stimulating, activating, amplifying, and/or propagating. In some such embodiments, further culturing is effected under conditions such that the viral vector integrates into the host genome of one or more cells. Incubation and/or engineering may be performed in a culture vessel, such as a unit, chamber, well, column, tube, set of tubes, valve, vial, petri dish, bag, or other container for culturing or incubating cells. In some embodiments, the composition or cell is incubated in the presence of a stimulating condition or a stimulating agent. Such conditions include those designed to induce proliferation, expansion, activation and/or survival of cells in a population to mimic antigen exposure and/or to elicit cells for genetic engineering (e.g., for the introduction of recombinant antigen receptors).
In some embodiments, the further incubation is performed at a temperature above room temperature, such as above or above about 25 ℃, such as typically above or above about 32 ℃, 35 ℃, or 37 ℃. In some embodiments, further incubation is effected at a temperature of or about 37 ℃ ± 2 ℃, such as at a temperature of or about 37 ℃.
In some embodiments, the further incubation is performed under conditions for stimulating and/or activating the cells, which may include one or more of the following: specific media, temperature, oxygen content, carbon dioxide content, time, agents (e.g., nutrients, amino acids, antibiotics, ions, and/or stimulatory factors (such as cytokines, chemokines, antigens, binding partners, fusion proteins, recombinant soluble receptors, and any other agents designed to activate cells)).
In some embodiments, the stimulation conditions or agents include one or more agents (e.g., stimulatory and/or auxiliary agents), such as ligands, capable of activating the intracellular signaling domain of the TCR complex. In some aspects, the agent turns on or initiates a TCR/CD3 intracellular signaling cascade in the T cell, such as an agent suitable for delivering a primary signal, e.g., to initiate activation of an ITAM-induced signal (e.g., those specific for a TCR component), and/or to promote a co-stimulatory signal (e.g., a co-stimulatory signal specific for a T cell co-stimulatory receptor), e.g., an anti-CD 3, anti-CD 28, or anti-41-BB (e.g., which is optionally bound to a solid support such as a bead) and/or one or more cytokines. The stimulators include anti-CD 3/anti-CD 28 beads (e.g., M-450 CD3/CD 28T cell expander and/or +.>Beads). Optionally, the amplification method may further comprise the step of adding an anti-CD 3 and/or anti-CD 28 antibody to the culture medium. In some embodiments, the stimulatory agent includes IL-2 and/or IL-15, for example, IL-2 concentration of at least about 10 units/mL.
In some embodiments, the stimulation conditions or stimulators include one or more agents (e.g., ligands) capable of activating the intracellular signaling domain of the TCR complex. In some aspects, the agent initiates or initiates a TCR/CD3 intracellular signaling cascade in the T cell. Such agents may include, for example, antibodies that bind to a solid support (e.g., beads), such as those antibodies that are specific for TCR components and/or co-stimulatory receptors (e.g., anti-CD 3, anti-CD 28); and/or one or more cytokines. Optionally, the amplification method may further comprise the step of adding anti-CD 3 and/or anti-CD 28 antibodies to the culture medium (e.g., at a concentration of at least about 0.5 ng/ml). In some embodiments, the stimulatory agent includes IL-2 and/or IL-15, for example, IL-2 concentration of at least about 10 units/mL, at least about 50 units/mL, at least about 100 units/mL, or at least about 200 units/mL.
The conditions may include one or more of the following: specific media, temperature, oxygen content, carbon dioxide content, time, agents (e.g., nutrients, amino acids, antibiotics, ions, and/or stimulatory factors (e.g., cytokines, chemokines, antigens, binding partners, fusion proteins, recombinant soluble receptors, and any other agents designed to activate cells)).
In some aspects, incubation is performed according to a variety of techniques, such as those described in the following documents: U.S. Pat. No. 6,040,1,77 to Riddell et al; klebaroff et al (2012) J Immunother.35 (9): 651-660; terakura et al (2012) blood.1:72-82; and/or Wang et al (2012) J Immunother35 (9): 689-701.
In some embodiments, the further incubation is performed in the same vessel or apparatus in which the contacting is performed. In some embodiments, further incubation is performed without rotation or centrifugation, which is typically performed after at least a portion of incubation performed under rotation (e.g., in combination with centrifugation or rotary inoculation). In some embodiments, the further incubation is performed outside the stationary phase, such as outside the chromatographic matrix, e.g., in solution.
In some embodiments, the further incubation is performed in a different container or device from the container or device in which the contacting is performed, such as by transferring (e.g., automatically transferring) the cell composition into a different container or device after the contacting with the viral particles and the agent.
In some embodiments, further culturing or incubation is performed, for example, to facilitate ex vivo amplification for greater than or greater than about 24 hours, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, or 14 days. In some embodiments, further culturing or incubating is performed for no more than 6 days, no more than 5 days, no more than 4 days, no more than 3 days, no more than 2 days, or no more than 24 hours.
In some embodiments, for example, the total duration of incubation with the stimulating agent is between or between about 1 hour and 96 hours, between 1 hour and 72 hours, between 1 hour and 48 hours, between 4 hours and 36 hours, between 8 hours and 30 hours, or between 12 hours and 24 hours, such as at least or about 6 hours, 12 hours, 18 hours, 24 hours, 36 hours, or 72 hours. In some embodiments, further incubation is performed at or about between 1 hour and 48 hours, between 4 hours and 36 hours, between 8 hours and 30 hours, or between 12 hours and 24 hours, inclusive.
In some embodiments, the methods provided herein do not include further culturing or incubation, e.g., do not include an ex vivo amplification step, or include a significantly shorter ex vivo amplification step.
In some embodiments, the stimulating agent is removed and/or isolated from the cells prior to engineering. In certain embodiments, the stimulating agent is removed and/or isolated from the cells after engineering. In certain embodiments, the stimulating agent is removed and/or isolated from the cells after engineering and prior to incubating the engineered cells, e.g., under conditions that promote proliferation and/or expansion. In certain embodiments, the stimulating agent is a stimulating agent as described in section I-B-1. In certain embodiments, the stimulating agent is removed and/or isolated from the cells as described in section I-B-2.
1. Carrier and method
In some embodiments, cells (e.g., T cells) are genetically engineered to express recombinant receptors. In some embodiments, engineering is by introducing one or more polynucleotides encoding recombinant receptors or portions or components thereof. Polynucleotides encoding recombinant receptors are also provided, as are vectors or constructs comprising such nucleic acids and/or polynucleotides.
In particular embodiments, the vector is a viral vector, a non-viral vector. In some cases, the vector is a viral vector, such as a retroviral vector, e.g., a lentiviral vector or a gamma retroviral vector.
In some embodiments, the polynucleotide encoding the recombinant receptor comprises at least one promoter operably linked to control expression of the recombinant receptor. In some examples, the polynucleotide comprises two, three, or more promoters operably linked to control expression of the recombinant receptor. In some embodiments, a polynucleotide may contain regulatory sequences (such as transcription and translation initiation and termination codons) specific for the type of host (e.g., bacterial, fungal, plant, or animal) into which the polynucleotide is introduced, as appropriate and in consideration of whether the polynucleotide is DNA-based or RNA-based. In some embodiments, the polynucleotide may contain regulatory/control elements such as promoters, enhancers, introns, polyadenylation signals, kozak consensus sequences, internal Ribosome Entry Sites (IRES), 2A sequences and splice acceptors or donors. In some embodiments, the polynucleotide may contain a non-native promoter operably linked to a nucleotide sequence encoding a recombinant receptor and/or one or more additional polypeptides. In some embodiments, the promoter is selected from the group consisting of RNA pol I, pol II, or pol III promoters. In some embodiments, the promoter is recognized by RNA polymerase II (e.g., CMV, SV40 early region, or adenovirus major late promoter). In another embodiment, the promoter is recognized by RNA polymerase III (e.g., a U6 or H1 promoter). In some embodiments, the promoter may be a non-viral promoter or a viral promoter, such as the Cytomegalovirus (CMV) promoter, the SV40 promoter, the RSV promoter, and promoters found in the long terminal repeat of murine stem cell viruses. Other known promoters are also contemplated.
In some embodiments, the promoter is or comprises a constitutive promoter. Exemplary constitutive promoters include, for example, simian virus 40 early promoter (SV 40), cytomegalovirus immediate early promoter (CMV), human ubiquitin C promoter (UBC), human elongation factor 1 alpha promoter (EF 1 alpha), mouse phosphoglycerate kinase 1 Promoter (PGK), and chicken beta-actin promoter (CAGG) coupled to CMV early enhancer. In some embodiments, the constitutive promoter is a synthetic or modified promoter. In some embodiments, the promoter is or comprises an MND promoter, which is a synthetic promoter containing the U3 region of the modified MoMuLV LTR with a myeloproliferative sarcoma virus enhancer (see Challita et al (1995) J.Virol.69 (2): 748-755). In some embodiments, the promoter is a tissue specific promoter. In another embodiment, the promoter is a viral promoter. In another embodiment, the promoter is a non-viral promoter. In some embodiments, exemplary promoters may include, but are not limited to, the human elongation factor 1 alpha (EF 1 alpha) promoter or modified forms thereof or MND promoters.
In another embodiment, the promoter is a regulated promoter (e.g., an inducible promoter). In some embodiments, the promoter is an inducible promoter or a repressible promoter. In some embodiments, the promoter comprises a Lac operator sequence, a tetracycline operator sequence, a galactose operator sequence, or a doxycycline operator sequence, or an analog thereof, or is capable of binding or recognition by a Lac repressor or a tetracycline repressor analog thereof. In some embodiments, the polynucleotide does not include regulatory elements, e.g., a promoter.
In some cases, the nucleic acid sequence encoding a recombinant receptor (e.g., chimeric Antigen Receptor (CAR)) contains a signal sequence encoding a signal peptide. Non-limiting illustrative examples of signal peptides include, for example, the GMCSFR alpha chain signal peptide shown in SEQ ID NO. 10 and encoded by the nucleotide sequence shown in SEQ ID NO. 9, the CD8 alpha signal peptide shown in SEQ ID NO. 11 or the CD33 signal peptide shown in SEQ ID NO. 12.
In some embodiments, the polynucleotide contains a nucleic acid sequence encoding one or more additional polypeptides (e.g., one or more markers and/or one or more effector molecules). In some embodiments, the one or more markers include a transduction marker, a surrogate marker, and/or a selection marker. Additional nucleic acid sequences introduced, for example, encoding one or more additional polypeptides include: nucleic acid sequences that can improve the efficacy of the therapy, such as by promoting the viability and/or function of the transferred cells; providing a genetically-tagged nucleic acid sequence for selecting and/or evaluating cells (e.g., assessing survival or localization in vivo); nucleic acid sequences that improve safety, for example, by making cells susceptible to in vivo negative selection, as described in: lupton s.d. et al, mol.and Cell biol.,11:6 (1991); and Riddell et al Human Gene Therapy3:319-338 (1992); see also WO 1992008796 and WO 1994028143 (describing the use of bifunctional selectable fusion genes derived from fusing a dominant positive selectable marker with a negative selectable marker), and U.S. patent No. 6,040,177.
In some embodiments, the marker is a transduction marker or an alternative marker. The transduction markers or surrogate markers can be used to detect cells into which a polynucleotide (e.g., a polynucleotide encoding a recombinant receptor) has been introduced. In some embodiments, the transduction marker may indicate or confirm modification to the cell. In some embodiments, the surrogate marker is a protein prepared to co-express with a recombinant receptor (e.g., CAR) on the cell surface. In particular embodiments, such surrogate markers are surface proteins that have been modified to have little or no activity. In certain embodiments, the surrogate markers are encoded by the same polynucleotide encoding the recombinant receptor. In some embodiments, the nucleic acid sequence encoding the recombinant receptor is operably linked to a nucleic acid sequence encoding a marker, optionally separated by an Internal Ribosome Entry Site (IRES) or a nucleic acid encoding a self-cleaving peptide or a ribosome jump-inducing peptide, such as the 2A sequence. In some cases, an extrinsic marker gene may be associated with an engineered cell for allowing detection or selection of the cell, and in some cases, may also be used to promote cell elimination and/or cell suicide.
Exemplary surrogate markers may include truncated forms of a cell surface polypeptide, such as truncated forms that are nonfunctional and do not transduce or are incapable of transducing a signal or are generally transduced by a full length form of the cell surface polypeptide, and/or are not internalized or are incapable of internalizing. Exemplary truncated cell surface The panel polypeptide includes truncated forms of a growth factor or other receptor, such as truncated human epidermal growth factor receptor 2 (tHER 2), truncated epidermal growth factor receptor (tEGFR, exemplary tEGFR sequences shown in SEQ ID NO:2 or 3), or Prostate Specific Membrane Antigen (PSMA) or modified forms thereof, such as truncated PSMA (tPSMA). In some aspects, the tggfr may contain a polypeptide derived from the antibody cetuximabOr other therapeutic anti-EGFR antibodies or binding molecules, which can be used to identify or select cells that have been engineered with the tgfr construct and the encoded foreign protein, and/or to eliminate or isolate cells expressing the encoded foreign protein. See U.S. patent No. 8,802,374 and Liu et al, nature biotech.2016, month 4; 34 (4):430-434). In some aspects, the marker (e.g., surrogate marker) includes all or part (e.g., truncated form) of CD34, NGFR, CD19, or truncated CD19 (e.g., truncated non-human CD 19). Exemplary polypeptides of truncated EGFR (e.g., tEGFR) comprise the amino acid sequence shown in SEQ ID NO:2 or 3 or an amino acid sequence exhibiting at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO:2 or 3.
In some embodiments, the label is or comprises a detectable protein, such as a fluorescent protein, e.g., green Fluorescent Protein (GFP), enhanced Green Fluorescent Protein (EGFP) (e.g., superfolder GFP (sfGFP)), red Fluorescent Protein (RFP) (e.g., tdTomato, mCherry, mStrawberry, asRed, dsRed, or DsRed 2), cyan Fluorescent Protein (CFP), blue-green fluorescent protein (BFP), enhanced Blue Fluorescent Protein (EBFP), and Yellow Fluorescent Protein (YFP), and variants thereof, including species variants, monomeric variants, codon optimized, stabilized, and/or enhanced variants of fluorescent protein. In some embodiments, the label is or comprises an enzyme (e.g., luciferase), the lacZ gene from E.coli, alkaline phosphatase, secreted Embryonic Alkaline Phosphatase (SEAP), chloramphenicol Acetyl Transferase (CAT). Exemplary luminescent reporter genes include luciferase (luc), beta-galactosidase, chloramphenicol Acetyl Transferase (CAT), beta-Glucuronidase (GUS), or variants thereof. In some aspects, the expression of an enzyme may be detected by adding a substrate that may be detected based on the expression and functional activity of the enzyme.
In some embodiments, the marker is a resistance marker or a selection marker. In some embodiments, the resistance marker or selectable marker is or comprises a polypeptide that confers resistance to an exogenous agent or drug. In some embodiments, the resistance marker or selectable marker is an antibiotic resistance gene. In some embodiments, the resistance marker or selectable marker is an antibiotic resistance gene that confers antibiotic resistance to mammalian cells. In some embodiments, the resistance marker or selectable marker is or comprises a puromycin resistance gene, a hygromycin resistance gene, a blasticidin resistance gene, a neomycin resistance gene, a geneticin resistance gene, or a bleomycin resistance gene, or a modified version thereof.
Any of the recombinant receptors and/or one or more additional polypeptides described herein may be encoded by one or more polynucleotides comprising one or more nucleic acid sequences encoding the recombinant receptors in any combination, orientation, or arrangement. For example, one, two, three, or more polynucleotides may encode one, two, three, or more different polypeptides (e.g., recombinant receptors or portions or components thereof) and/or one or more additional polypeptides (e.g., markers and/or effector molecules). In some embodiments, a polynucleotide comprises a nucleic acid sequence encoding a recombinant receptor (e.g., CAR) or a portion or component thereof, and a nucleic acid sequence encoding one or more additional polypeptides. In some embodiments, one vector or construct contains a nucleic acid sequence encoding a recombinant receptor (e.g., CAR) or a portion or component thereof, and a separate vector or construct contains a nucleic acid sequence encoding one or more additional polypeptides. In some embodiments, the nucleic acid sequence encoding the recombinant receptor and the nucleic acid sequence encoding one or more additional polypeptides are operably linked to two different promoters. In some embodiments, the nucleic acid encoding the recombinant receptor is present upstream of the nucleic acid encoding the one or more additional polypeptides. In some embodiments, the nucleic acid encoding the recombinant receptor is present downstream of the nucleic acid encoding the one or more additional polypeptides.
In certain instances, a polynucleotide comprises a nucleic acid sequence encoding two or more different polypeptide chains, e.g., a recombinant receptor and one or more additional polypeptides, e.g., markers and/or effector molecules. In some embodiments, nucleic acid sequences encoding two or more different polypeptide chains (e.g., a recombinant receptor and one or more additional polypeptides) are present in two separate polynucleotides. For example, two separate polynucleotides are provided, and each may be transferred or introduced separately into a cell for expression in the cell. In some embodiments, the nucleic acid sequence encoding the marker and the nucleic acid sequence encoding the recombinant receptor are present or inserted at different locations within the genome of the cell. In some embodiments, the nucleic acid sequence encoding the marker and the nucleic acid sequence encoding the recombinant receptor are operably linked to two different promoters.
In some embodiments, such as those in which the polynucleotide comprises first and second nucleic acid sequences, the coding sequences encoding each of the different polypeptide chains may be operably linked to the same or different promoters. In some embodiments, the nucleic acid molecule may contain a promoter that drives expression of two or more different polypeptide chains. In some embodiments, such nucleic acid molecules may be polycistronic (bicistronic or tricistronic), see, e.g., U.S. patent No. 6,060,273. In some embodiments, the nucleic acid sequence encoding the recombinant receptor and the nucleic acid sequence encoding one or more additional polypeptides are operably linked to the same promoter and optionally separated by an Internal Ribosome Entry Site (IRES) or a nucleic acid encoding a self-cleaving peptide or a ribosome jump-inducing peptide (e.g., 2A element). For example, an exemplary tag and optional ribosome jump sequence can be any of those disclosed in PCT publication No. WO 2014031687.
In some embodiments, the transcriptional unit may be engineered to contain an IRES bicistronic unit that allows the gene product (e.g., encoding a recombinant receptor and additional polypeptide) to be co-expressed by information from a single promoter. Alternatively, in some cases, a single promoter may direct expression of RNAs that contain two or three genes (e.g., encoding a tag and encoding a recombinant receptor) separated from each other by a sequence encoding a self-cleaving peptide (e.g., a 2A sequence) or a protease recognition site (e.g., furin) in a single Open Reading Frame (ORF). Thus, the ORF encodes a single polypeptide that is processed into separate proteins during translation (in the case of 2A) or post-translationally. In some cases, peptides such as T2A may cause ribosomes to skip synthesis of peptide bonds at the C-terminus of the 2A element (ribosome skipping), resulting in separation between the 2A sequence end and the next peptide downstream (see, e.g., de Felipe, genetic Vaccines and Ther.2:13 (2004) and de Felipe et al Traffic 5:616-626 (2004)). Various 2A elements are known. Examples of 2A sequences that may be used in the methods and systems disclosed herein include, but are not limited to, 2A sequences from: foot and mouth disease virus (F2A, e.g., SEQ ID NO: 8), equine rhinitis A virus (E2A, e.g., SEQ ID NO: 7), leptopetalum album beta tetrad virus (Thosea asigna virus) (T2A, e.g., SEQ ID NO:1 or 4), and porcine teschovirus (porcine teschovirus) -1 (P2A, e.g., SEQ ID NO:5 or 6), as described in U.S. patent publication No. 20070116690.
In some embodiments, the polynucleotide encoding the recombinant receptor and/or additional polypeptide is contained in a vector or may be cloned into one or more vectors. In some embodiments, one or more vectors may be used to transform or transfect a host cell, e.g., for an engineered cell. Exemplary vectors include vectors designed for introduction, propagation and amplification or for expression or both, such as plasmids and viral vectors. In some aspects, the vector is an expression vector, e.g., a recombinant expression vector. In some embodiments, standard recombinant DNA techniques may be used to prepare recombinant expression vectors.
In some embodiments, the vector may be a series of vectors as follows: pUC series (Fermentas Life Sciences), pBluescript series (Stratagene, lahough, california), pET series (Novagen, madison, wis.), pGEX series (Pharmacia Biotech, uppsala, sweden) or pEX series (Clontech, pa Luo Aotu, california). In some cases, phage vectors such as λg10, λgt11, λ ZapII (Stratagene), λembl4, and λnm1149 may also be used. In some embodiments, plant expression vectors may be used and include pBI01, pBI101.2, pBI101.3, pBI121, and pBIN19 (Clontech). In some embodiments, animal expression vectors include pEUK-Cl, pMAM and pMAMneo (Clontech).
In some embodiments, the polynucleotide encoding the recombinant receptor and/or one or more additional polypeptides is introduced into a composition comprising cultured cells, such as by retroviral transduction, transfection, or transformation.
In some embodiments, the vector is a viral vector, such as a retroviral vector. In some embodiments, polynucleotides encoding recombinant receptors and/or one or more additional polypeptides are introduced into cells by retroviruses or lentiviral vectors or by transposons (see, e.g., baum et al (2006) Molecular Therapy: the Journal of the American Society of Gene therapy.13:1050-1063; frecha et al (2010) Molecular Therapy18:1748-1757; and Hackett et al (2010) Molecular Therapy 18:674-683).
In some embodiments, the vector comprises a viral vector, such as a retrovirus or lentivirus, a non-viral vector, or a transposon, such as the sleeping beauty transposon system; vectors derived from simian virus 40 (SV 40), adenovirus, adeno-associated virus (AAV); lentiviral vectors or retroviral vectors, such as gamma-retroviral vectors, retroviral vectors derived from Moloney (Moloney) murine leukemia virus (MoMLV), myeloproliferative sarcoma virus (MPSV), murine embryonic stem cell virus (MESV), murine Stem Cell Virus (MSCV), spleen Focus Forming Virus (SFFV) or adeno-associated virus (AAV).
In some embodiments, electroporation is used to introduce ONE or more polynucleotides into T cells (see, e.g., chicaybam et al, (2013) PLoS ONE 8 (3): e60298; and Van Tedeloo et al (2000) Gene Therapy 7 (16): 1431-1437). In some embodiments, the recombinant nucleic acid is transferred into T cells by transposition (see, e.g., manuri et al (2010) Hum Gene Ther 21 (4): 427-437; shalma et al (2013) Molec Ther Nucl Acids, e74; and Huang et al (2009) Methods Mol Biol 506: 115-126). Other methods of introducing and expressing genetic material (e.g., polynucleotides and/or vectors) in immune cells include calcium phosphate transfection (e.g., as described in Current Protocols in Molecular Biology, john Wiley & Sons, new york.n.y.), protoplast fusion, cationic liposome-mediated transfection; tungsten particle-promoted microprojectile bombardment (Johnston, nature,346:776-777 (1990)); and strontium phosphate DNA co-precipitation (Brash et al, mol. Cell biol.,7:2031-2034 (1987)) and other methods such as described in international patent application publication No. WO 2014055668 and U.S. patent No. 7,446,190.
In some embodiments, one or more polynucleotides or vectors encoding recombinant receptors and/or one or more additional polypeptides may be introduced into a cell (e.g., a T cell) during or after amplification. For example, the introduction of one or more polynucleotides or one or more vectors may be performed using any suitable retroviral vector. The resulting genetically engineered cells can then be freed from the initial stimulus (e.g., anti-CD 3/anti-CD 28 stimulus) and subsequently stimulated in the presence of a second type of stimulus (e.g., by a recombinant receptor introduced de novo). The second type of stimulus may include an antigen stimulus in the form of a peptide/MHC molecule, a cognate (cross-linked) ligand of a genetically introduced receptor (e.g., the natural antigen and/or ligand of a CAR), or any ligand (e.g., an antibody) that binds directly within the framework of a new receptor (e.g., by recognizing a constant region within the receptor). See, e.g., cheadle et al, "Chimeric antigen receptors for T-cell based therapy" Methods Mol biol.2012;907:645-66; or Barrett et al, chimeric Antigen Receptor Therapy for Cancer Annual Review of Medicine, volume 65:333-347 (2014).
In some cases, vectors may be used that do not require activating cells (e.g., T cells). In some such cases, the cells may be selected and/or transduced prior to activation. Thus, the cells may be engineered before or after culturing the cells, and in some cases, at least a portion of the culturing.
a. Viral vector particles
In some embodiments, one or more polynucleotides are introduced into the cell using recombinant infectious viral particles, such as, for example, vectors derived from simian virus 40 (SV 40), adenovirus, adeno-associated virus (AAV). In some embodiments, one or more polynucleotides are introduced into T cells using a recombinant lentiviral vector or a retroviral vector (e.g., a gamma-retroviral vector) (see, e.g., koste et al (2014) Gene Therapy 2014, month 3. Doi:10.1038/gt.2014.25; carlens et al (2000) Exp Hematol 28 (10): 1137-46; alonso-Camino et al (2013) Mol Ther Nucl Acids 2, e93; park et al, trends Biotechnol.2011, month 11 (11): 550-557).
In some embodiments, the vector is a retroviral vector. In some embodiments, the retroviral vector has a Long Terminal Repeat (LTR), e.g., a retroviral vector derived from moloney murine leukemia virus (MoMLV), myeloproliferative sarcoma virus (MPSV), murine embryonic stem cell virus (MESV), murine Stem Cell Virus (MSCV), spleen Focus Forming Virus (SFFV), or adeno-associated virus (AAV). Most retroviral vectors are derived from murine retroviruses. In some embodiments, retroviruses include those derived from any avian or mammalian cell source. Retroviruses are often amphotropic, meaning that they are capable of infecting host cells of several species, including humans. In one embodiment, the gene to be expressed replaces retroviral gag, pol and/or env sequences. A number of exemplary retroviral systems have been described (e.g., U.S. Pat. Nos. 5,219,740, 6,207,453, 5,219,740; miller and Rosman (1989) BioTechniques 7:980-990; miller, A.D. (1990) Human Gene Therapy 1:5-14; scarpa et al (1991) Virology 180:849-852; burns et al (1993) Proc. Natl. Acad. Sci. USA 90:8033-8037; and Boris-Lawrie and Temin (1993) Cur. Opin. Genet. Development.3:102-109).
Methods of lentiviral transduction are known. Exemplary methods are described, for example, in the following documents: wang et al (2012) J.Immunother35 (9): 689-701; cooper et al (2003) blood.101:1637-1644; verhoeyen et al (2009) Methods Mol biol.506:97-114; cavalieri et al (2003) blood.102 (2): 497-505.
In some embodiments, the viral vector particles contain a genome derived from a retroviral genome-based vector (e.g., derived from a lentiviral genome-based vector). In some aspects of the provided viral vectors, a heterologous nucleic acid encoding a recombinant receptor (e.g., an antigen receptor, such as a CAR) is contained and/or located between the 5'ltr and 3' ltr sequences of the vector genome.
In some embodiments, the viral vector genome is a lentiviral genome, such as an HIV-1 genome or an SIV genome. For example, lentiviral vectors have been generated by attenuating virulence genes multiple times, e.g., genes env, vif, vpu and nef can be deleted, making the vector safer for therapeutic purposes. Lentiviral vectors are known. See Naldini et al, (1996 and 1998); zufferey et al, (1997); dull et al, 1998, U.S. Pat. Nos. 6,013,516; and 5,994,136). In some embodiments, these viral vectors are plasmid-based or virus-based and are configured to carry a base sequence for incorporation of foreign nucleic acids for selection and for transferring the nucleic acids into host cells. Known lentiviruses can be readily obtained from a custody institution or collection such as the american type culture collection ("ATCC"; university of marasas, virginia (University blvd)), 10801, 20110-2209) or isolated from known sources using conventional techniques.
Non-limiting examples of lentiviral vectors include those derived from lentiviruses, such as human immunodeficiency virus 1 (HIV-1), HIV-2, simian Immunodeficiency Virus (SIV), human T-lymphotropic virus 1 (HTLV-1), HTLV-2, or equine infectious anemia virus (E1 AV). For example, lentiviral vectors have been generated by attenuating HIV virulence genes multiple times, e.g., deleting the genes env, vif, vpr, vpu and nef, making the vector safer for therapeutic purposes. Lentiviral vectors are known in the art, see Naldini et al, (1996 and 1998); zufferey et al, (1997); dull et al, 1998, U.S. Pat. Nos. 6,013,516; and 5,994,136). In some embodiments, these viral vectors are plasmid-based or virus-based and are configured to carry a base sequence for incorporation of foreign nucleic acids for selection and for transferring the nucleic acids into host cells. Known lentiviruses can be readily obtained from a custody institution or collection such as the american type culture collection ("ATCC"; university of marasas, virginia (University blvd)), 10801, 20110-2209) or isolated from known sources using conventional techniques.
In some embodiments, the viral genome vector may contain sequences of the 5 'and 3' LTRs of a retrovirus (e.g., lentivirus). In some aspects, the viral genome construct may contain sequences from the 5 'and 3' ltrs of the lentivirus, and in particular may contain the R and U5 sequences from the 5 'ltrs of the lentivirus, as well as the inactivated or self-inactivated 3' ltrs from the lentivirus. The LTR sequence may be an LTR sequence of any lentivirus from any species. For example, they may be LTR sequences from HIV, SIV, FIV or BIV. Typically, the LTR sequence is an HIV LTR sequence.
In some embodiments, the nucleic acid of a viral vector (e.g., an HIV viral vector) lacks additional transcriptional units. The vector genome may contain an inactivated or self-inactivated 3' LTR (Zufferey et al J Virol 72:9873,1998; miyoshi et al J Virol 72:8150, 1998). For example, a deletion in the U3 region of the 3' LTR of the nucleic acid used to generate viral vector RNA may be used to generate a self-inactivating (SIN) vector. This deletion can then be transferred to the 5' LTR of proviral DNA during reverse transcription. Self-inactivating vectors typically have deletions from the enhancer and promoter sequences of the 3 'Long Terminal Repeat (LTR) that are copied into the 5' LTR during vector integration. In some embodiments, sufficient sequence may be deleted, including removal of the TATA box, to eliminate transcriptional activity of the LTR. This can prevent the generation of full-length vector RNA in transduced cells. In some aspects, the U3 element of the 3' LTR contains deletions of its enhancer sequence, TATA box, sp1, and NF-. Kappa.B sites. Due to self-inactivating the 3'LTR, provirus generated after entry and reverse transcription contains an inactivated 5' LTR. This may improve safety by reducing the risk of mobilizing the vector genome and the effect of the LTR on nearby cell promoters. The self-inactivating 3' LTR may be constructed by any method known in the art. In some embodiments, this does not affect the vector titer or the in vitro or in vivo properties of the vector.
Optionally, the U3 sequence from the lentiviral 5' LTR may be replaced with a promoter sequence (e.g., a heterologous promoter sequence) in the viral construct. This may increase the titer of the virus recovered from the packaging cell line. Enhancer sequences may also be included. Any enhancer/promoter combination that increases expression of the viral RNA genome in the packaging cell line may be used. In one example, a CMV enhancer/promoter sequence is used (U.S. patent No. 5,385,839 and U.S. patent No. 5,168,062).
In certain embodiments, the risk of insertional mutagenesis can be minimized by constructing the retroviral vector genome (e.g., lentiviral vector genome) to be integration defective. A variety of approaches can be used to generate non-integrated vector genomes. In some embodiments, one or more mutations may be engineered into the integrase component of the pol gene such that it encodes a protein with an inactive integrase. In some embodiments, the vector genome itself may be modified to prevent integration by, for example, mutating or deleting one or both attachment sites, or to render the 3' ltr Proximal Polypurine Tract (PPT) nonfunctional by deletion or modification. In some embodiments, non-genetic pathways may be used; these pathways include pharmacological agents that inhibit one or more functions of the integrase. These methods are not mutually exclusive; that is, more than one of the methods may be used at a time. For example, both the integrase and the attachment site may be nonfunctional, or both the integrase and the PPT site may be nonfunctional, or both the attachment site and the PPT site may be nonfunctional, or both may be nonfunctional. Such methods and viral vector genomes are known and available (see Philpott and Thresher, human Gene Therapy 18:483,2007; engelman et al J Virol 69:2729,1995; brown et al J Virol 73:9011 (1999); WO 2009/076524; mcWilliams et al J Virol 77:11150,2003; powell and Levin J Virol 70:5288, 1996).
In some embodiments, the vector contains sequences for propagation in a host cell (e.g., a prokaryotic host cell). In some embodiments, the nucleic acid of the viral vector contains one or more origins of replication for propagation in prokaryotic cells (e.g., bacterial cells). In some embodiments, vectors comprising a prokaryotic origin of replication may also contain genes whose expression confers a detectable or selectable marker, such as resistance.
Viral vector genomes are typically constructed in the form of plasmids that can be transfected into packaging or production cell lines. Any of a variety of known methods can be used to produce retroviral particles whose genome contains an RNA copy of the viral vector genome. In some embodiments, at least two components are involved in preparing a viral-based gene delivery system: first, the packaging plasmid, comprising the structural proteins and the enzymes necessary for the production of the viral vector particles, and second, the viral vector itself, i.e. the genetic material to be transferred. Biosafety protection can be incorporated in the design of one or both of these components.
In some embodiments, the packaging plasmid may contain all retroviral (e.g., HIV-1) proteins except for the envelope proteins (Naldini et al, 1998). In other embodiments, the viral vector may lack additional viral genes (such as those associated with virulence, e.g., vpr, vif, vpu and nef and/or Tat (the primary transactivator of HIV)). In some embodiments, a lentiviral vector (e.g., an HIV-based lentiviral vector) comprises only the genes of three parental viruses: gag, pol and rev, which reduces or eliminates the possibility of reconstitution of the wild-type virus by recombination.
In some embodiments, the viral vector genome is introduced into a packaging cell line that contains all components required to package viral genomic RNA transcribed from the viral vector genome into viral particles. Alternatively, the viral vector genome may comprise one or more genes encoding viral components in addition to one or more sequences of interest (e.g., recombinant nucleic acids). However, in some aspects, to prevent replication of the genome in the target cell, endogenous viral genes required for replication are removed and provided separately in the packaging cell line.
In some embodiments, the packaging cell line is transfected with one or more plasmid vectors containing components necessary to produce the particles. In some embodiments, plasmids containing the viral vector genome (including the LTR, cis-acting packaging sequence, and sequence of interest, i.e., nucleic acid encoding an antigen receptor (e.g., CAR)), are used; and one or more helper plasmids encoding viral enzymes and/or structural components (e.g., gag, pol, and/or rev). In some embodiments, various genetic components of retroviral vector particles are isolated using a variety of vectors. In some such embodiments, providing the packaging cell with a separate vector reduces the likelihood of recombination events that might otherwise generate replication competent viruses. In some embodiments, a single plasmid vector with all retroviral components may be used.
In some embodiments, retroviral vector particles (e.g., lentiviral vector particles) are pseudotyped to increase the transduction efficiency of a host cell. For example, in some embodiments, retroviral vector particles (e.g., lentiviral vector particles) are pseudotyped with VSV-G glycoprotein, which provides a broad cell host range, thereby expanding the cell types that can be transduced. In some embodiments, packaging cell lines are transfected with a plasmid or polynucleotide encoding a non-native envelope glycoprotein to include, for example, a amphotropic, amphotropic or amphotropic envelope, such as sindbis virus envelope, GALV or VSV-G.
In some embodiments, the packaging cell line provides components, including viral regulatory proteins and structural proteins, required for trans-action in packaging viral genomic RNA into lentiviral vector particles. In some embodiments, the packaging cell line may be any cell line capable of expressing a lentiviral protein and producing a functional lentiviral vector particle. In some aspects, suitable packaging cell lines include 293 (ATCC CCL X), 293T, heLA (ATCC CCL 2), D17 (ATCC CCL 183), MDCK (ATCC CCL 34), BHK (ATCC CCL-10) and Cf2Th (ATCC CRL 1430) cells.
In some embodiments, the packaging cell line stably expresses one or more viral proteins. For example, in some aspects, packaging cell lines containing gag, pol, rev and/or other structural genes but without LTRs and packaging components may be constructed. In some embodiments, packaging cell lines may be transiently transfected with nucleic acid molecules encoding one or more viral proteins, viral vector genomes containing nucleic acid molecules encoding heterologous proteins, and/or nucleic acids encoding envelope glycoproteins.
In some embodiments, the viral vector and packaging plasmid and/or helper plasmid are introduced into the packaging cell line by transfection or infection. Packaging cell lines produce viral vector particles containing viral vector genomes. Methods for transfection or infection are well known. Non-limiting examples include calcium phosphate, DEAE-dextran, and lipofection methods, electroporation, and microinjection.
When the recombinant plasmid and retroviral LTRs and packaging sequences are introduced into a particular cell line (e.g., by calcium phosphate precipitation), the packaging sequences may allow for RNA transcription of the recombinant plasmid to be packaged into viral particles, which may then be secreted into the culture medium. In some embodiments, the recombinant retrovirus-containing medium is then collected, optionally concentrated, and used for gene transfer. For example, in some aspects, after co-transfection of the packaging plasmid and transfer vector into the packaging cell line, the viral vector particles are recovered from the culture medium and stepwise adjusted by standard methods used by those skilled in the art.
In some embodiments, retroviral vectors, such as lentiviral vectors, may be produced in packaging cell lines (e.g., the exemplary HEK 293T cell line) by introducing a plasmid to allow for the production of lentiviral particles. In some embodiments, the packaging cells are transfected and/or contain polynucleotides encoding gag and pol, as well as polynucleotides encoding recombinant receptors (e.g., antigen receptors, such as CARs). In some embodiments, the packaging cell line is optionally and/or additionally transfected with and/or contains a polynucleotide encoding a rev protein. In some embodiments, the packaging cell line is optionally and/or additionally transfected with and/or contains a polynucleotide encoding a non-native envelope glycoprotein (e.g., VSV-G). In some such embodiments, the cell supernatant contains recombinant lentiviral vectors that can be recovered and stepwise adjusted approximately two days after transfection of the cells (e.g., HEK 293T cells).
The recovered and/or produced retroviral vector particles can be used to transduce target cells using the methods as described. Once in the target cell, the viral RNA is reverse transcribed, enters the nucleus and is stably integrated into the host genome. Expression of a recombinant protein (e.g., an antigen receptor such as a CAR) can be detected one or two days after viral RNA integration.
In some embodiments, provided methods relate to methods of transducing cells by contacting (e.g., incubating) a cell composition comprising a plurality of cells with a viral particle. In some embodiments, the cells to be transfected or transduced are or comprise primary cells obtained from the subject, e.g., cells enriched and/or selected from the subject.
In some embodiments, the concentration of cells to be transduced in the composition is from 1.0x10 5 Individual cells/mL to 1.0x10 8 Individual cells/mL or from about 1.0x10 5 Individual cells/mL to about 1.0x10 8 Individual cells/mL, e.g., water stop or about at least or about 1.0x10 5 Individual cells/mL, 5X10 5 Individual cells/mL, 1X10 6 Individual cells/mL, 5X10 6 Individual cells/mL, 1X10 7 Individual cells/mL, 5X10 7 Individual cells/mL or 1x10 8 Individual cells/mL.
In some embodiments, the viral particles are provided in a ratio of copies of the viral vector particles or units of Infection (IU) thereof to the total number of cells to be transduced (IU/cells). For example, in some embodiments, the viral particles are present during contact as viral vector particles at or about or at least about 0.5, 1, 2, 3, 4, 5, 10, 15, 20, 30, 40, 50, or 60IU per cell.
In some embodiments, the titer of the viral vector particles is at or about 1x10 6 IU/mL and 1x10 8 IU/mL, e.g., at or about 5x10 6 IU/mL and 5x10 7 Between IU/mL, e.g. at least 6x10 6 IU/mL、7x10 6 IU/mL、8x10 6 IU/mL、9x10 6 IU/mL、1x10 7 IU/mL、2x10 7 IU/mL、3x10 7 IU/mL、4x10 7 IU/mL or 5x10 7 IU/mL。
In some embodiments, transduction may be achieved at a multiplicity of infection (MOI) of less than 100, such as typically less than 60, 50, 40, 30, 20, 10, 5 or less.
In some embodiments, the methods involve contacting or incubating the cells with a viral particle. In some embodiments, the contacting is performed for 30 minutes to 72 hours, such as 30 minutes to 48 hours, 30 minutes to 24 hours, or 1 hour to 24 hours, for example at least or about 30 minutes, 1 hour, 2 hours, 6 hours, 12 hours, 24 hours, 36 hours, or more.
In some embodiments, the contacting is performed in solution. In some embodiments, the cells and virus particles are contacted in the following volumes: from 0.5mL to 500mL or from about 0.5mL to about 500mL, such as from or from about 0.5mL to 200mL, 0.5mL to 100mL, 0.5mL to 50mL, 0.5mL to 10mL, 0.5mL to 5mL, 5mL to 500mL, 5mL to 200mL, 5mL to 100mL, 5mL to 50mL, 5mL to 10mL, 10mL to 500mL, 10mL to 200mL, 10mL to 100mL, 10mL to 50mL, 50mL to 500mL, 50mL to 200mL, 50mL to 100mL, 100mL to 500mL, 100mL to 200mL or 200mL to 500mL.
In certain embodiments, the input cells are treated, incubated, or contacted with particles comprising binding molecules that bind to or recognize recombinant receptors encoded by viral DNA.
In some embodiments, incubating the cells with the viral vector particles results in or produces an output composition comprising cells transduced with the viral vector particles.
b. Non-viral vectors
In some embodiments, the recombinant nucleic acid is transferred into T cells by electroporation (see, e.g., chicaybam et al, (2013) PLoS ONE 8 (3): e60298; and Van Tedeloo et al (2000) Gene Therapy7 (16): 1431-1437). In some embodiments, the recombinant nucleic acid is transferred into T cells by transposition (see, e.g., manuri et al (2010) Hum Gene Ther 21 (4): 427-437; shalma et al (2013) Molec Ther Nucl Acids, e74; and Huang et al (2009) Methods Mol Biol 506: 115-126). Other methods of introducing and expressing genetic material in immune cells include calcium phosphate transfection (e.g., as described in Current Protocols in Molecular Biology, john Wiley & Sons, new york.n.y.), protoplast fusion, cationic liposome-mediated transfection; tungsten particle-promoted microprojectile bombardment (Johnston, nature,346:776-777 (1990)); and strontium phosphate DNA co-precipitation (Brash et al, mol. Cell biol.,7:2031-2034 (1987)).
Other methods and vectors for transferring nucleic acids encoding recombinant products are those described, for example, in International patent application publication No. WO 2014055668 and U.S. Pat. No. 7,446,190.
In some embodiments, the recombinant nucleic acid is transferred into a T cell via a transposon. Transposons (transposable elements) are DNA moveable segments that can move from one locus to another within the genome. These elements move through a conservative "cut-and-paste" mechanism: transposases catalyze excision of transposons from their original location and promote their re-integration elsewhere in the genome. If the transposase is provided by another transposase gene, the element lacking the transposase can be mobilized. Thus, transposons can be used to incorporate foreign DNA into the host genome without the use of viral transduction systems. Examples of transposons suitable for use with mammalian cells (e.g., human primary leukocytes) include, but are not limited to, sleeping Beauty (Sleeping beautyy) and PiggyBacs.
Transposon-based transfection is a two-component system consisting of a transposase and a transposon. In some embodiments, the system comprises a transposon engineered to comprise foreign DNA (also referred to herein as cargo DNA), such as a gene encoding a recombinant receptor, flanked by inverted repeat/direct repeat (IR/DR) sequences recognized by a concomitant transposase. In some embodiments, the non-viral plasmid encodes a transposase under the control of a promoter. Transfection of the plasmid into the host cell results in transient expression of the transposase, whereby during the initial period following transfection, the transposase is expressed at a sufficient level to integrate the transposon into genomic DNA. In some embodiments, the transposase itself is not integrated into genomic DNA, and thus the expression of the transposase decreases over time. In some embodiments, the transposase expression is expressed by the host cell at a level sufficient to allow integration of the corresponding transposon for the following time: less than about 4 hours, less than about 8 hours, less than about 12 hours, less than about 24 hours, less than about 2 days, less than about 3 days, less than about 4 days, less than about 5 days, less than about 6 days, less than about 7 days, less than about 2 weeks, less than about 3 weeks, less than about 4 weeks, less than about weeks, or less than about 8 weeks. In some embodiments, cargo DNA introduced into the host genome is not subsequently removed from the host genome, at least because the host does not express an endogenous transposase capable of excising cargo DNA.
Sleeping Beauty (SB) is a synthetic member of the Tc/1-water arm superfamily of transposons, reconstructed from dormant elements found in the salmonidae fish genome. SB transposon-based transfection is a two-component system consisting of a transposase and a transposon containing an inverted repeat/direct repeat (IR/DR) sequence that results in precise integration into the TA dinucleotide. Transposons are designed with the expression cassette of interest flanked by IR/DR. SB transposases bind to specific binding sites located on sleeping beauty transposon IR. SB transposases mediate integration of transposons, which are mobile elements encoding a cargo sequence flanked on both sides by inverted terminal repeats having catalytic enzyme (SB) binding sites. Stable expression is obtained when SB inserts the gene sequence into the vertebrate chromosome at the TA target dinucleotide by a cut-and-paste mechanism. This system has been used to engineer a variety of vertebrate cell types, including human primary peripheral blood leukocytes. In some embodiments, the cell is contacted with, incubated with, and/or treated with a SB transposon that comprises a cargo gene (e.g., a gene encoding a recombinant receptor or CAR) flanked by SB IR sequences. In particular embodiments, the cells to be transfected are contacted with, incubated with, and/or treated with a plasmid comprising a SB transposon that comprises a cargo gene (e.g., a gene encoding a CAR) flanked by SB IR sequences. In certain embodiments, the plasmid further comprises a gene encoding an SB transposase flanked by no SB IR sequences.
PiggyBac (PB) is another transposon system that may be used to integrate cargo DNA into the genomic DNA of a host (e.g., a human). The PB transposase recognizes PB transposon-specific Inverted Terminal Repeats (ITRs) located at both ends of the transposon, and efficiently removes the content from the original site and integrates the content into TTAA chromosomal site. The PB transposon system enables mobilization of the gene of interest between two ITRs in the PB vector into the target genome. The PB system has been used to engineer a variety of vertebrate cell types, including human primary cells. In some embodiments, the cells to be transfected are contacted with, incubated with, and/or treated with a PB transposon that comprises a cargo gene (e.g., a gene encoding a CAR) flanked by PB IR sequences. In particular embodiments, the cells to be transfected are contacted with, incubated with, and/or treated with a plasmid comprising a PB transposon that comprises a cargo gene (e.g., a gene encoding a CAR) flanked by PB IR sequences. In certain embodiments, the plasmid further comprises a gene encoding an SB transposase flanked by no PB IR sequences.
In some embodiments, the various elements of transposon/transposase used in the subject methods, such as one or more SB or PB vectors, can be produced by standard methods of restriction enzyme cleavage, ligation, and molecular cloning. One approach for constructing the subject vector includes the following steps. First, a purified nucleic acid fragment containing the desired component nucleotide sequence as well as foreign sequences is cleaved with a restriction endonuclease from an initial source (e.g., a vector comprising a transposase gene). The fragments containing the desired nucleotide sequence are then separated from the different sized unwanted fragments using conventional separation methods (e.g., by agarose gel electrophoresis). The desired fragments are excised from the gel and ligated together in the appropriate configuration to produce a circular nucleic acid or plasmid containing the desired sequences (e.g., sequences corresponding to the various elements of the subject vector as described above). The thus constructed circular molecule is then amplified in a prokaryotic host (e.g., E.coli) if desired. The procedures involved in these steps, cleavage, plasmid construction, cell transformation and plasmid generation are well known to those skilled in the art, and the enzymes required for restriction and ligation are commercially available. (see, e.g., R.Wu editions, methods in Enzymology, volume 68, academic Press, N.Y. (1979); T.Maniatis, E.F.Fritsch and J.Sambrook, molecular Cloning: A Laboratory Manual, cold Spring Harbor Laboratory Press, cold Spring Harbor, N.Y. (1982); catalog nos. 1982-83,New England Biolabs,Inc; catalog nos. 1982-83,Bethesda Research Laboratories,Inc. Examples of how to construct vectors for use in the subject methods are provided in the experimental section below. Preparation of representative sleeping beauty transposon systems is also disclosed in WO 98/40510 and WO 99/25817).
In some embodiments, transduction with a transposon containing a cargo DNA sequence flanked by inverted repeat/direct repeat (IR/DR) sequences recognized by a transposase is performed with a plasmid comprising a transposase gene and a plasmid comprising a transposon. In certain embodiments, the cargo DNA sequence encodes a heterologous protein, such as a recombinant T cell receptor or CAR. In some embodiments, the plasmid comprises a transposase and a transposon. In some embodiments, the transposase is under the control of a ubiquitous promoter or any promoter suitable for driving expression of the transposase in a target cell. Ubiquitous promoters include, but are not limited to, EF1a, CMB, SV40, PGK1, ubc, human beta-actin, CAG, TRE, UAS, ac, caMKIIa, and U6. In some embodiments, the cargo DNA comprises a selection cassette that allows for selection of cells that stably integrate the cargo DNA into genomic DNA. Suitable selection boxes include, but are not limited to, selection boxes encoding: kanamycin resistance gene, spectinomycin resistance gene, streptomycin resistance gene, ampicillin resistance gene, carbenicillin resistance gene, hygromycin resistance gene, bleomycin resistance gene, erythromycin resistance gene and polymyxin B resistance gene.
In some embodiments, the component for transduction with a transposon (e.g., a plasmid comprising SB transposase and SB transposon) is introduced into the target cell. Any convenient protocol may be employed, wherein the protocol may introduce the system components into the target cells in vitro or in vivo, depending on the location of the target cells. For example, where the target cell is an isolated cell, the system may be introduced directly into the cell under cell culture conditions that allow for viability of the target cell, e.g., by using standard transformation techniques. Such techniques include, but are not necessarily limited to: viral infection, transformation, conjugation, protoplast fusion, electroporation, particle gun technology, calcium phosphate precipitation, direct microinjection, viral vector delivery, and the like. The choice of method generally depends on the type of cell to be transformed and the environment in which the transformation occurs (i.e., in vitro, ex vivo, or in vivo). A general discussion of these methods can be found in Ausubel et al, short Protocols in Molecular Biology, 3 rd edition, wiley & Sons, 1995.
In some embodiments, the SB transposon and the source of SB transposase are introduced into a target cell of a multicellular organism (e.g., a mammal or human) under conditions sufficient to excise the inverted repeat flanking nucleic acid from the vector carrying the transposon and subsequently integrate the excised nucleic acid into the genome of the target cell. Some embodiments further comprise the step of ensuring that the requisite transposase activity is present in the target cell along with the introduced transposon. Depending on the structure of the transposon vector itself, i.e. whether the vector comprises a region encoding a product having transposase activity, the method may further comprise introducing a second vector encoding the requisite transposase activity into the target cell.
In some embodiments, the amount of transposon-containing vector nucleic acid and the amount of vector nucleic acid encoding a transposase introduced into the cell are sufficient to provide the desired excision and insertion of the transposon nucleic acid into the target cell genome. Thus, the amount of vector nucleic acid introduced should provide a sufficient amount of transposase activity and a sufficient copy number of the nucleic acid desired to be inserted into the target cell. The amount of vector nucleic acid introduced into the target cell varies depending on the efficiency of the particular introduction protocol employed (e.g., the particular ex vivo administration protocol employed).
Once the vector DNA has been combined with the requisite transposase into the target cell, the region of the vector nucleic acid flanked by inverted repeats (i.e., the vector nucleic acid located between the inverted repeats recognized by the sleeping American transposase) is excised from the vector by the provided transposase and inserted into the genome of the target cell. Thus, after introducing the vector DNA into the target cell, a transposase-mediated excision of the exogenous nucleic acid carried by the vector is then performed and inserted into the genome of the target cell. In particular embodiments, the vector is integrated into the genome of at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least 7%, at least 8%, at least 9%, at least 10%, at least 15%, or at least 20% of cells transfected with the SB transposon and/or SB transposase. In some embodiments, the integration of the nucleic acid into the target cell genome is stable, i.e., the vector nucleic acid remains present in the target cell genome for more than a transient period of time, and a portion of the chromosomal genetic material is transferred to the progeny of the target cell.
In certain embodiments, transposons are used to integrate nucleic acids (i.e., polynucleotides) of various sizes into the genome of a target cell. In some embodiments, the DNA size inserted into the genome of the target cell using the subject methods ranges from about 0.1kb to 200kb, from about 0.5kb to 100kb, from about 1.0kb to about 8.0kb, from about 1.0 to about 200kb, from about 1.0 to about 10kb, from about 10kb to about 50kb, from about 50kb to about 100kb, or from about 100kb to about 200kb. In some embodiments, the DNA size inserted into the target cell genome using the subject methods ranges from about 1.0kb to about 8.0kb. In some embodiments, the DNA size inserted into the target cell genome using the subject methods ranges from about 1.0kb to about 200kb. In particular embodiments, the DNA size inserted into the target cell genome using the subject methods ranges from about 1.0kb to about 8.0kb.
D. Cell culture and/or expansion
In some embodiments, provided methods include one or more steps for incubating cells (e.g., incubating cells under conditions that promote proliferation and/or expansion). In some embodiments, following the step of genetic engineering (e.g., introduction of a recombinant polypeptide into a cell by transduction or transfection), the cell is incubated under conditions that promote proliferation and/or expansion. In certain embodiments, the cells are incubated under stimulating conditions and the cells are incubated after transduction or transfection with a recombinant polynucleotide (e.g., a polynucleotide encoding a recombinant receptor). In some embodiments, the culturing produces one or more culturing compositions that enrich for T cells.
In certain embodiments, one or more compositions of enriched T cells (including stimulated and transduced T cells) (separate compositions of such cd4+ and cd8+ T cells) are incubated prior to formulating the cells, e.g., under conditions that promote proliferation and/or expansion. In some aspects, methods of culturing (e.g., for promoting proliferation and/or amplification) include methods as provided herein in section I-F. In certain embodiments, after one or more T cell enriched compositions have been engineered (e.g., transduced or transfected), the one or more compositions are incubated. In certain embodiments, one or more of the compositions is an engineered composition. In certain embodiments, one or more of the engineered compositions have been previously cryogenically frozen and stored and thawed prior to incubation.
In certain embodiments, one or more compositions of engineered T cells are or comprise two separate compositions of enriched T cells. In particular embodiments, two separate compositions of enriched T cells introduced with recombinant receptor (e.g., CAR) are separately incubated under conditions that promote proliferation and/or expansion of the cells, e.g., two separate compositions of enriched T cells selected, isolated, and/or enriched from the same biological sample. In some embodiments, the conditions are stimulating conditions. In certain embodiments, the two separate compositions comprise compositions enriched for cd4+ T cells (e.g., engineered cd4+ T cells that have incorporated nucleic acid encoding a recombinant receptor and/or express a recombinant receptor). In particular embodiments, the two separate compositions include compositions enriched for cd8+ T cells (e.g., engineered cd8+ T cells introduced with nucleic acid encoding a recombinant receptor and/or expressing a recombinant receptor). In some embodiments, the two separate compositions enriched for cd4+ T cells and enriched for cd8+ T cells (e.g., engineered cd4+ T cells and engineered cd8+ T cells) are separately incubated, e.g., under conditions that promote proliferation and/or expansion. In some embodiments, a single composition enriched for T cells is incubated. In certain embodiments, the single composition is a composition enriched for cd4+ T cells. In some embodiments, the single composition is a composition enriched for cd4+ and cd8+ T cells that have been combined from separate compositions prior to incubation.
In some embodiments, a composition of enriched cd4+ T cells (e.g., engineered cd4+ T cells), e.g., incubated under conditions that promote proliferation and/or expansion, comprises at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or is about 100% cd4+ T cells. In some embodiments, the composition comprises at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or at least about 100% of cd4+ T cells that express the recombinant receptor and/or that have been transduced or transfected with a recombinant polynucleotide encoding the recombinant receptor. In certain embodiments, the cultured cd4+ T cell-enriched composition comprises less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1%, or less than 0.01% cd8+ T cells, and/or is free of cd8+ T cells, and/or is free or substantially free of cd8+ T cells.
In some embodiments, a composition of enriched cd8+ T cells (e.g., engineered cd8+ T cells), e.g., incubated under conditions that promote proliferation and/or expansion, comprises at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or is about 100% cd8+ T cells. In particular embodiments, the composition comprises at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or at least about 100% of cd8+ T cells expressing the recombinant receptor and/or that have been transduced or transfected with a recombinant polynucleotide encoding the recombinant receptor. In certain embodiments, the composition of enriched cd8+ T cells incubated under stimulation conditions comprises less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1%, or less than 0.01% of cd4+ T cells, and/or is free or substantially free of cd4+ T cells.
In some embodiments, separate compositions enriched for cd4+ and cd8+ T cells (e.g., separate compositions of engineered cd4+ and engineered cd8+ T cells) are combined into a single composition and incubated, for example, under conditions that promote proliferation and/or expansion. In certain embodiments, the separately incubated compositions enriched for cd4+ and enriched for cd8+ T cells are combined into a single composition after incubation has been performed and/or completed. In certain embodiments, separate compositions enriched for cd4+ and cd8+ T cells (e.g., separate compositions of engineered cd4+ and engineered cd8+ T cells) are incubated separately, e.g., under conditions that promote proliferation and/or expansion.
In some embodiments, cells (e.g., engineered cells) are cultured in a volume of medium that is, is about, or is at least 100mL, 200mL, 300mL, 400mL, 500mL, 600mL, 700mL, 800mL, 900mL, 1,000mL, 1,200mL, 1,400mL, 1,600mL, 1,800mL, 2,000mL, 2,200mL, or 2,400 mL. In some embodiments, cells are incubated at an initial volume, which is later adjusted to a different volume. In certain embodiments, the volume is adjusted later during incubation. In certain embodiments, the volume increases from the initial volume during incubation. In certain embodiments, the volume is increased as the cells achieve density during incubation. In certain embodiments, the initial volume is at or about 500mL.
In certain embodiments, the volume increases from the initial volume as the cells achieve a density or concentration during incubation. In certain embodiments, the volume is increased when the cells achieve the following densities and/or concentrations:is, is about, or is at least 0.1x10 6 Individual cells/ml, 0.2x10 6 Individual cells/ml, 0.4x10 6 Individual cells/ml, 0.6x10 6 Individual cells/ml, 0.8x10 6 Individual cells/ml, 1X10 6 Individual cells/ml, 1.2x10 6 Individual cells/ml, 1.4x10 6 Individual cells/ml, 1.6x10 6 Individual cells/ml, 1.8x10 6 Individual cells/ml, 2.0x10 6 Individual cells/ml, 2.5x10 6 Individual cells/ml, 3.0x10 6 Individual cells/ml, 3.5x10 6 Individual cells/ml, 4.0x10 6 Individual cells/ml, 4.5x10 6 Individual cells/ml, 5.0x10 6 Individual cells/ml, 6X10 6 Individual cells/ml, 8x10 6 Individual cells/ml, or 10x10 6 Individual cells/ml. In some embodiments, when the cell is implemented as, at least, or about 0.6x10 6 At a density and/or concentration of individual cells/ml, the volume increases from the initial volume. In some embodiments, the density and/or concentration is of living cells in culture. In certain embodiments, the volume is increased when the cells achieve the following densities and/or concentrations: is, is about, or is at least 0.1x10 6 Living cells/ml, 0.2x10 6 Living cells/ml, 0.4x10 6 Living cells/ml, 0.6x10 6 Living cells/ml, 0.8x10 6 Living cells/ml, 1X10 6 Living cells/ml, 1.2x10 6 Living cells/ml, 1.4x10 6 Living cells/ml, 1.6x10 6 Living cells/ml, 1.8x10 6 Living cells/ml, 2.0x10 6 Living cells/ml, 2.5x10 6 Living cells/ml, 3.0x10 6 Living cells/ml, 3.5x10 6 Living cells/ml, 4.0x10 6 Living cells/ml, 4.5x10 6 Living cells/ml, 5.0x10 6 Living cells/ml, 6X10 6 Living cells/ml, 8X10 6 Individual living cells/ml, or 10x10 6 Each living cell/ml. In some embodiments, when a living cell is realized as, is at least or about 0.6x10 6 The volume is increased from the initial volume at the density and/or concentration of individual living cells/ml. In some embodiments, the density and/or concentration of cells or living cells may be determined or monitored during incubation, such as by using a method as described,including optical methods including Digital Holographic Microscopy (DHM) or Differential Digital Holographic Microscopy (DDHM).
In some embodiments, the cells achieve a certain density and/or concentration, and the volume increases, or increases by about or at least 100mL, 200mL, 300mL, 400mL, 500mL, 600mL, 700mL, 800mL, 900mL, 1,000mL, 1,200mL, 1,400mL, 1,600mL, 1,800mL, 2,000mL, 2,200mL, or 2,400mL. In some embodiments, the volume is increased by 500mL. In certain embodiments, the volume is increased to the following volume: is, is about or at least 500mL, 600mL, 700mL, 800mL, 900mL, 1,000mL, 1,200mL, 1,400mL, 1,600mL, 1,800mL, 2,000mL, 2,200mL, or 2,400mL. In certain embodiments, the volume increases to a volume of 1,000 ml. In certain embodiments, the volume increases at the following rate: 5mL, 10mL, 20mL, 25mL, 30mL, 40mL, 50mL, 60mL, 70mL, 75mL, 80mL, 90mL, or 100mL for, at least, or about every 1,2, 3, 4, 5, 6, 7, 8, 9, or 10 minutes. In certain embodiments, the rate is at or about 50mL per 8 minutes.
In some embodiments, the composition of enriched T cells (e.g., engineered T cells) is incubated under conditions that promote proliferation and/or expansion. In some embodiments, such conditions may be designed to induce proliferation, expansion, activation, and/or survival of cells in a population. In particular embodiments, the stimulation conditions may include one or more of the following: specific media, temperature, oxygen content, carbon dioxide content, time, agents (e.g., nutrients, amino acids, antibiotics, ions, and/or stimulatory factors (e.g., cytokines, chemokines, antigens, binding partners, fusion proteins, recombinant soluble receptors, and any other agents designed to promote growth, division, and/or expansion of cells)).
In some embodiments, the incubation is performed under conditions that generally include a temperature suitable for the growth of primary immune cells (e.g., human T lymphocytes), such as at least about 25 degrees celsius, typically at least about 30 degrees celsius, and typically at or about 37 degrees celsius. In some embodiments, the T cell enriched composition is incubated at a temperature of 25 ℃ to 38 ℃, such as 30 ℃ to 37 ℃, for example, at or about 37 ℃ ± 2 ℃. In some embodiments, incubation is performed for a period of time until the culturing (e.g., incubating or expanding) produces the desired or threshold density, concentration, number, or dose of cells. In some embodiments, incubation is performed for a period of time until the culture (e.g., incubation or expansion) produces a desired or threshold density, concentration, number, or dose of living cells. In some embodiments, the incubation is greater than or greater than about or for about or 24 hours, 48 hours, 72 hours, 96 hours, 5 days, 6 days, 7 days, 8 days, 9 days, or more. In some embodiments, the density, concentration and/or number or dose of cells may be determined or monitored during incubation, such as by using methods as described, including optical methods, including Digital Holographic Microscopy (DHM) or Differential Digital Holographic Microscopy (DDHM).
In some embodiments, the stimulating agent is removed and/or isolated from the cells prior to incubation. In certain embodiments, the stimulatory agent is removed and/or isolated from the engineered cells after engineering and prior to incubating the cells, e.g., under conditions that promote proliferation and/or expansion. In some embodiments, the stimulating agent is a stimulating agent as described herein, for example, in section I-B-1. In certain embodiments, the stimulating agent is removed and/or isolated from the cells as described herein (e.g., in section I-B-2).
In certain embodiments, a composition of enriched T cells (e.g., engineered T cells) is incubated in the presence of one or more cytokines (e.g., a separate composition of engineered cd4+ T cells and engineered cd8+ T cells). In certain embodiments, the one or more cytokines are recombinant cytokines. In certain embodiments, the one or more cytokines are human recombinant cytokines. In certain embodiments, one or more cytokines bind and/or are capable of binding to receptors expressed by and/or endogenous to T cells. In certain embodiments, the one or more cytokines are or include members of the 4-alpha-helix bundle family of cytokines. In some embodiments, members of the 4- α -helix bundle family of cytokines include, but are not limited to, interleukin 2 (IL-2), interleukin 4 (IL-4), interleukin 7 (IL-7), interleukin 9 (IL-9), interleukin 12 (IL-12), interleukin 15 (IL-15), granulocyte colony-stimulating factor (G-CSF), and granulocyte macrophage colony-stimulating factor (GM-CSF). In some embodiments, one or more cytokines is or includes IL-15. In certain embodiments, the one or more cytokines is or includes IL-7. In certain embodiments, the one or more cytokines are or include recombinant IL-2.
In certain embodiments, the composition enriched for cd4+ T cells (e.g., engineered cd4+ T cells) is incubated with recombinant IL-2. In some embodiments, incubating a composition enriched for cd4+ T cells (e.g., engineered cd4+ T cells) in the presence of recombinant IL-2 increases the probability or likelihood that cd4+ T cells of the composition will continue to survive, grow, expand, and/or activate during the incubation step and throughout the process. In some embodiments, incubating the composition enriched for cd4+ T cells (e.g., engineered cd4+ T cells) in the presence of recombinant IL-2 increases the probability and/or likelihood that an output composition enriched for cd4+ T cells (e.g., engineered cd4+ T cells suitable for cell therapy) will be produced from the composition enriched for cd4+ T cells by at least 0.5%, at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least 7%, at least 8%, at least 9%, at least 10%, at least 11%, at least 12%, at least 13%, at least 14%, at least 15%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 100%, or at least 200% cd4+ as compared to an alternative and/or exemplary method of incubating the composition enriched for cd4+ T cells in the presence of recombinant IL-2.
In some embodiments, the cells (e.g., a separate composition of engineered cd4+ T cells and engineered cd8+ T cells) are incubated with cytokines, e.g., recombinant human cytokines, at a concentration of between 1IU/ml and 2,000IU/ml, between 10IU/ml and 100IU/ml, between 50IU/ml and 500IU/ml, between 100IU/ml and 200IU/ml, between 500IU/ml and 1400IU/ml, between 250IU/ml and 500IU/ml, or between 500IU/ml and 2,500IU ml.
In some embodiments, the enriched T cell composition (e.g., the separate compositions of engineered CD4+ T cells and engineered CD8+ T cells) is incubated with recombinant IL-2, e.g., human recombinant IL-2, at a concentration of between 2IU/ml and 500IU/ml, between 10IU/ml and 250IU/ml, between 100IU/ml and 500IU/ml, or between 100IU/ml and 400IU/ml. In particular embodiments, the composition enriched in T cells is incubated with IL-2 at a concentration of at or about 50IU/ml, 75IU/ml, 100IU/ml, 125IU/ml, 150IU/ml, 175IU/ml, 200IU/ml, 225IU/ml, 250IU/ml, 300IU/ml, or 400IU/ml. In some embodiments, the T cell enriched composition is incubated with recombinant IL-2 at a concentration of 200 IU/ml. In some embodiments, the T cell enriched composition is a cd4+ T cell enriched composition, such as an engineered cd4+ T cell composition. In particular embodiments, the T cell enriched composition is a cd8+ T cell enriched composition, such as an engineered cd8+ T cell composition.
In some embodiments, a composition enriched for T cells (e.g., a separate composition of engineered CD4+ T cells and CD8+ T cells) is incubated with IL-7, e.g., human recombinant IL-7, at a concentration of between 10IU/ml and 5,000IU/ml, between 500IU/ml and 2,000IU/ml, between 600IU/ml and 1,500IU/ml, between 500IU/ml and 2,500IU/ml, between 750IU/ml and 1,500IU/ml, or between 1,000IU/ml and 2,000 IU/ml. In particular embodiments, the composition enriched in T cells is incubated with IL-7 at a concentration of at or about 100IU/ml, 200IU/ml, 300IU/ml, 400IU/ml, 500IU/ml, 600IU/ml, 700IU/ml, 800IU/ml, 900IU/ml, 1,000IU/ml, 1,200IU/ml, 1,400IU/ml, or 1,600IU/ml. In some embodiments, in the concentration of or about 1,200IU/ml recombinant IL-7 in the presence of cultured cells. In some embodiments, the T cell enriched composition is a composition enriched for cd4+ T cells (e.g., engineered cd4+ T cells).
In some embodiments, a composition of enriched T cells (e.g., a separate composition of engineered CD4+ T cells and CD8+ T cells) is incubated with IL-15, e.g., human recombinant IL-15, at a concentration of between 0.1IU/ml and 200IU/ml, between 1IU/ml and 50IU/ml, between 5IU/ml and 25IU/ml, between 25IU/ml and 50IU/ml, between 5IU/ml and 15IU/ml, or between 10IU/ml and 100 IU/ml. In particular embodiments, the enriched T cell composition is incubated with IL-15 at a concentration of at or about 1IU/ml, 2IU/ml, 3IU/ml, 4IU/ml, 5IU/ml, 6IU/ml, 7IU/ml, 8IU/ml, 9IU/ml, 10IU/ml, 11IU/ml, 12IU/ml, 13IU/ml, 14IU/ml, 15IU/ml, 20IU/ml, 25IU/ml, 30IU/ml, 40IU/ml, 50IU/ml, 100IU/ml, or 200IU/ml. In a particular embodiment, the T cell enriched composition is incubated with recombinant IL-15 at a concentration of 20 IU/ml. In some embodiments, the T cell enriched composition is a composition enriched for cd4+ T cells (e.g., engineered cd4+ T cells). In particular embodiments, the T cell enriched composition is a cd8+ T cell enriched (e.g., engineered cd8+ T cell) composition.
In certain embodiments, the composition enriched for cd8+ T cells (e.g., engineered cd8+ T cells) is incubated in the presence of IL-2 and/or IL-15 (e.g., in an amount as described). In certain embodiments, the composition enriched for CD4+ T cells (e.g., engineered CD4+ T cells) is incubated in the presence of IL-2, IL-7, and/or IL-15 (e.g., in the amounts described). In some embodiments, IL-2, IL-7 and/or IL-15 is recombinant. In certain embodiments, IL-2, IL-7 and/or IL-15 is human. In particular embodiments, the one or more cytokines are or include human recombinant IL-2, IL-7, and/or IL-15.
In certain embodiments, the incubation is performed in a closed system. In certain embodiments, the incubation is performed in a closed system under sterile conditions. In certain embodiments, incubation is performed in the same closed system as one or more steps of the provided system. In some embodiments, the T cell enriched composition is removed from the closed system and placed in and/or connected to a bioreactor for cultivation. Examples of suitable bioreactors for incubation include, but are not limited to, GE Xuri W25, GE Xuri W5, sartorius BioSTAT RM 20|50, finesse SmartRocker bioreactor systems, and Pall XRS bioreactor systems. In some embodiments, the cells are perfused and/or mixed using a bioreactor during at least a portion of the incubating step.
In some embodiments, cells cultured in a closed, connected bioreactor and/or under control of the bioreactor undergo faster expansion during culturing than cells cultured without the bioreactor (e.g., cells cultured under static conditions (e.g., without mixing, rocking, movement, and/or perfusion). In some embodiments, cells cultured in a closed, connected bioreactor and/or under control of the bioreactor reach or achieve a threshold expansion, cell count, and/or density within 14 days, 10 days, 9 days, 8 days, 7 days, 6 days, 5 days, 4 days, 3 days, 2 days, 60 hours, 48 hours, 36 hours, 24 hours, or 12 hours. In some embodiments, cells grown in a closed, linked bioreactor and/or under control of the bioreactor reach or achieve a threshold expansion, cell count and/or density of at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 100%, at least 150%, at least 1-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, as compared to cells grown in an exemplary and/or alternative process that do not grow cells in a closed, linked bioreactor and/or under control of the bioreactor.
In some embodiments, the mixing is or includes rocking and/or motion. In some cases, the bioreactor may be subject to motion or rocking, which in some aspects may increase oxygen transfer. Moving the bioreactor may include, but is not limited to, rotation along a horizontal axis, rotation along a vertical axis, rocking movement along a horizontal axis of tilt (or incorporated) of the bioreactor, or any combination thereof. In some embodiments, at least a portion of the incubation is performed with rocking. The rocking speed and rocking angle can be adjusted to achieve the desired agitation. In some embodiments, the rocking angle is 20 °, 19 °, 18 °, 17 °, 16 °, 15 °, 14 °, 13 °, 12 °, 11 °, 10 °, 9 °, 8 °, 7 °, 6 °, 5 °, 4 °, 3 °, 2 °, or 1 °. In certain embodiments, the rocking angle is between 6 ° and 16 °. In other embodiments, the rocking angle is between 7 ° and 16 °. In other embodiments, the rocking angle is between 8 ° and 12 °. In some embodiments, the rocking rate is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 1 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40rpm. In some embodiments, the rocking rate is between 4rpm and 12rpm, for example between 4rpm and 6rpm, and includes an end value.
In some embodiments, the bioreactor is maintained at a temperature at or near 37 ℃ and at or near 5% CO 2 Horizontal, and has a stable air flow as follows: is about or at least 0.01L/min, 0.05L/min, 0.1L/min, 0.2L/min, 0.3L/min, 0.4L/min, 0.5L/min, 1.0L/min, 1.5L/min or 2.0L/min or greater than 2.0L/min. In certain embodiments, at least a portion of the incubation is performed under perfusion conditions, such as at a rate of 290 ml/day, 580 ml/day, and/or 1160 ml/day (e.g., depending on the timing associated with initiation of incubation and/or the density of the incubated cells). In some embodiments, at least a portion of the cell culture expansion is performed with a rocking motion, such as at an angle between 5 ° and 10 ° (e.g., 6 °), at a constant rocking speed, such as a speed between 5 and 15RPM (e.g., 6RPM or 10 RPM).
In some embodiments, the at least a portion of the incubating step is performed under constant perfusion (e.g., slow steady rate perfusion). In some embodiments, the perfusion is or includes an outflow of liquid (e.g., spent media) and an inflow of fresh media. In certain embodiments, the perfusion is replaced with fresh medium with spent medium. In some embodiments, at least a portion of the incubation is performed under perfusion with the following steady rates: about or at least 100 ml/day, 200 ml/day, 250 ml/day, 275 ml/day, 290 ml/day, 300 ml/day, 350 ml/day, 400 ml/day, 450 ml/day, 500 ml/day, 550 ml/day, 575 ml/day, 580 ml/day, 600 ml/day, 650 ml/day, 700 ml/day, 750 ml/day, 800 ml/day, 850 ml/day, 900 ml/day, 950 ml/day, 1000 ml/day, 1100 ml/day, 1160 ml/day, 1200 ml/day, 1400 ml/day, 1600 ml/day, 1800 ml/day, 2000 ml/day, 2200 ml/day, or 2400 ml/day.
In particular embodiments, incubation is initiated without perfusion and perfusion is initiated after a set and/or predetermined amount of time (e.g., at or about or at least 12 hours, 24 hours, 36 hours, 48 hours, 60 hours, or 72 hours or more than 72 hours after initiation of incubation). In certain embodiments, perfusion begins when the density or concentration of cells reaches a set or predetermined density or concentration. In some embodiments, perfusion begins when the cultured cells reach the following densities or concentrations: is, is about or at least 0.1x10 6 Individual cells/ml, 0.2x10 6 Individual cells/ml, 0.4x10 6 Individual cells/ml, 0.6x10 6 Individual cells/ml, 0.8x10 6 Individual cells/ml, 1X10 6 Individual cells/ml, 1.2x10 6 Individual cells/ml, 1.4x10 6 Individual cells/ml, 1.6x10 6 Individual cells/ml, 1.8x10 6 Individual cells/ml, 2.0x10 6 Individual cells/ml, 2.5x10 6 Individual cells/ml, 3.0x10 6 Individual cells/ml, 3.5x10 6 Individual cells/ml, 4.0x10 6 Individual cells/ml, 4.5x10 6 Individual cells/ml, 5.0x10 6 Individual cells/ml, 6X10 6 Individual cells/ml, 8x10 6 Individual cells/ml or 10x10 6 Individual cells/ml. In certain embodiments, perfusion begins when the density or concentration of living cells reaches a set or predetermined density or concentration. In some embodiments, perfusion begins when cultured living cells reach the following densities or concentrations: is, is about or at least 0.1x10 6 Living cells/ml, 0.2x10 6 Living cells/ml, 0.4x10 6 Living cells/ml, 0.6x10 6 Living cells/ml, 0.8x10 6 Living cells/ml, 1X10 6 Living cells/ml, 1.2x10 6 Living cells/ml, 1.4x10 6 Living cells/ml, 1.6x10 6 Living cells/ml, 1.8x10 6 Living cells/ml, 2.0x10 6 Living cells/ml, 2.5x10 6 Living cells/ml, 3.0x10 6 Living cells/ml, 3.5x10 6 Living cells/ml, 4.0x10 6 Living cells/ml, 4.5x10 6 Living cells/ml, 5.0x10 6 Living cells/ml, 6X10 6 Living cells/ml, 8X10 6 Individual living cells/ml or 10x10 6 Each living cell/ml.
In certain embodiments, perfusion is performed at different rates during incubation. For example, in some embodiments, the rate of perfusion depends on the density and/or concentration of the cultured cells. In certain embodiments, the rate of perfusion is increased when the cells reach a set or predetermined density or concentration. The perfusion rate may be changed during incubation, for example, from a steady perfusion rate to an increased steady perfusion rate, once, twice, three times, four times, five times, more than ten times, more than 15 times, more than 20 times, more than 25 times, more than 50 times, or more than 100 times. In some embodiments, the steady perfusion rate is increased when the cells reach a set or predetermined cell density or concentration as follows: is, is about or at least 0.6x10 6 Individual cells/ml, 0.8x10 6 Individual cells/ml, 1X10 6 Individual cells/ml, 1.2x10 6 Individual cells/ml, 1.4x10 6 Individual cells/ml, 1.6x10 6 Individual cells/ml, 1.8x10 6 Individual cells/ml, 2.0x10 6 Individual cells/ml, 2.5x10 6 Individual cells/ml, 3.0x10 6 Individual cells/ml, 3.5x10 6 Individual cells/ml, 4.0x10 6 Individual cells/ml, 4.5x10 6 Individual cells/ml, 5.0x10 6 Individual cells/ml, 6X10 6 Individual cells/ml, 8x10 6 Individual cells/ml or 10x10 6 Individual cells/ml. In some embodiments, the steady perfusion rate is increased when the cells reach a viable cell density or concentration set or predetermined as follows: is, is about or at least 0.6x10 6 Living cells/ml, 0.8x10 6 Living cells/ml, 1X10 6 Living cells/ml, 1.2x10 6 Living cells/ml, 1.4x10 6 Living cells/ml, 1.6x10 6 Living cells/ml, 1.8x10 6 Living cells/ml, 2.0x10 6 Living cells/ml, 2.5x10 6 Living cells/ml, 3.0x10 6 Living cells/ml, 3.5x10 6 Living cells/ml, 4.0x10 6 Living cells/ml, 4.5x10 6 Living cells/ml, 5.0x10 6 Living cells/ml, 6X10 6 Living cells/ml, 8X10 6 Individual living cells/ml or 10x10 6 Each living cell/ml. In some embodiments, the density and/or concentration of cells or living cells during incubation (e.g., under perfusion) may be determined or monitored, such as by using methods as described, including optical methods, including Digital Holographic Microscopy (DHM) or Differential Digital Holographic Microscopy (DDHM).
In some embodiments, incubation is initiated without perfusion and perfusion is initiated when the density or concentration of cells reaches a set or predetermined density or concentration. In some embodiments, when the density or concentration of cells reaches a set or predetermined density or concentration, perfusion begins at the following rate: is, is about or at least 100 ml/day, 200 ml/day, 250 ml/day, 275 ml/day, 290 ml/day, 300 ml/day, 350 ml/day, 400 ml/day, 450 ml/day, 500 ml/day, 550 ml/day, 575 ml/day, 580 ml/day, 600 ml/day, 650 ml/day, 700 ml/day, 750 ml/day, 800 ml/day, 850 ml/day, 900 ml/day, 950 ml/day, 1000 ml/day, 1100 ml/day, 1160 ml/day, 1200 ml/day, 1400 ml/day, 1600 ml/day, 1800 ml/day, 2000 ml/day, 2200 ml/day, or 2400 ml/day. In some embodiments, perfusion begins when cultured cells or cultured living cells reach the following densities or concentrations: is, is about or at least 0.1x10 6 Individual cells/ml, 0.2x10 6 Individual cells/ml, 0.4x10 6 Individual cells/ml, 0.6x10 6 Individual cells/ml, 0.8x10 6 Individual cells/ml, 1X10 6 Individual cells/ml, 1.2x10 6 Individual cells/ml, 1.4x10 6 Individual cells/ml, 1.6x10 6 Individual cells/ml, 1.8x10 6 Individual cells/ml, 2.0x10 6 Individual cells/ml, 2.5x10 6 Individual cells/ml, 3.0x10 6 Individual cells/ml, 3.5x10 6 Individual cells/ml, 4.0x10 6 Individual cells/ml, 4.5x10 6 Individual cells/ml, 5.0x10 6 Individual cells/ml, 6X10 6 Individual cells/ml, 8x10 6 Individual cells/ml or 10x10 6 Individual cells/ml.
In certain embodiments, at least a portion of the incubation is performed at a rate of perfusion and when the density or concentration of cells reaches a set or predetermined density or concentrationAt the concentration, the perfusion rate was increased to the following rate: is, is about or at least 100 ml/day, 200 ml/day, 250 ml/day, 275 ml/day, 290 ml/day, 300 ml/day, 350 ml/day, 400 ml/day, 450 ml/day, 500 ml/day, 550 ml/day, 575 ml/day, 580 ml/day, 600 ml/day, 650 ml/day, 700 ml/day, 750 ml/day, 800 ml/day, 850 ml/day, 900 ml/day, 950 ml/day, 1000 ml/day, 1100 ml/day, 1160 ml/day, 1200 ml/day, 1400 ml/day, 1600 ml/day, 1800 ml/day, 2000 ml/day, 2200 ml/day, or 2400 ml/day. In some embodiments, perfusion begins when cultured cells or cultured living cells reach the following densities or concentrations: is, is about or at least 0.1x10 6 Individual cells/ml, 0.2x10 6 Individual cells/ml, 0.4x10 6 Individual cells/ml, 0.6x10 6 Individual cells/ml, 0.8x10 6 Individual cells/ml, 1X10 6 Individual cells/ml, 1.2x10 6 Individual cells/ml, 1.4x10 6 Individual cells/ml, 1.6x10 6 Individual cells/ml, 1.8x10 6 Individual cells/ml, 2.0x10 6 Individual cells/ml, 2.5x10 6 Individual cells/ml, 3.0x10 6 Individual cells/ml, 3.5x10 6 Individual cells/ml, 4.0x10 6 Individual cells/ml, 4.5x10 6 Individual cells/ml, 5.0x10 6 Individual cells/ml, 6X10 6 Individual cells/ml, 8x10 6 Individual cells/ml or 10x10 6 Individual cells/ml. In some embodiments, perfusion is performed when cells are incubated at the following volumes: is, is about or at least 300mL, 400mL, 500mL, 600mL, 700mL, 800mL, 900mL or 1000mL. In some embodiments, the volume is 1000mL.
In certain embodiments, incubation is initiated without perfusion or at a rate, and when the density or concentration of cells reaches, is about or at least 0.61x10 6 At a concentration of individual cells/ml, the perfusion rate is increased to, about or at least 290 ml/day. In certain embodiments, when the cells are incubated in a volume of, about, or at least 1000mL, when the density or concentration of the cells reaches, is, or is, about, or at least 0.61x10 6 Cells are perfused at a rate of, or at least about 290 ml/day at a concentration of individual cells/ml. In some embodiments, when the density or concentration of cells reaches, is About or at least 0.81x10 6 At a concentration of individual cells/ml, the perfusion rate is increased to, about or at least 580 ml/day. In certain embodiments, when the density or concentration of cells reaches, is about or at least 1.01x10 6 At a concentration of individual cells/ml, the infusion rate was increased to, about or at least 1160 ml/day. In some embodiments, when the density or concentration of cells reaches, is about or at least 1.2x10 6 At a concentration of individual cells/ml, the infusion rate was increased to, about or at least 1160 ml/day.
In aspects of the provided embodiments, the perfusion rate is determined by assessing the density and/or concentration of cells or assessing the density and/or concentration of living cells during incubation, including timing at which perfusion is initiated or increased as described herein and above. In some embodiments, the density and/or concentration of cells may be determined using methods as described, including optical methods, including Digital Holographic Microscopy (DHM) or Differential Digital Holographic Microscopy (DDHM).
In some embodiments, the composition of enriched cells, such as engineered T cells (e.g., engineered cd4+ T cells or engineered cd8+ T cells), is incubated in the presence of a surfactant. In certain embodiments, incubating the cells of the composition reduces the amount of shear stress that may occur during incubation, for example, due to mixing, rocking, movement, and/or perfusion. In particular embodiments, the composition of enriched T cells (e.g., engineered T cells, e.g., engineered cd4+ T cells or engineered cd8+ T cells) is incubated with a surfactant, and at least 50%, at least 60%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or at least 99.9% of the T cells are viable, e.g., are viable and/or do not undergo necrosis, programmed cell death, or apoptosis, for a period of 1 day, 2 days, 4 days, 5 days, 6 days, 7 days, or more than 7 days, or at least 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, or more than 7 days after completion of incubation. In particular embodiments, the composition of enriched T cells (e.g., engineered T cells, e.g., engineered cd4+ T cells or engineered cd8+ T cells) is incubated in the presence of a surfactant, and less than 50%, less than 40%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1%, or less than 0.01% of the cells do not undergo cell death, e.g., programmed cell death, apoptosis, and/or necrosis, as a result of shear or shear induced stress.
In certain embodiments, the composition of enriched T cells (e.g., engineered T cells, e.g., engineered cd4+ T cells or engineered cd8+ T cells) is incubated in the presence of the following amount of surfactant: between 0.1 and 10.0. Mu.l/ml, between 0.2 and 2.5. Mu.l/ml, between 0.5 and 5. Mu.l/ml, between 1 and 3. Mu.l/ml or between 2 and 4. Mu.l/ml. In some embodiments, the composition of enriched T cells (e.g., engineered T cells, e.g., engineered cd4+ T cells or engineered cd8+ T cells) is incubated in the presence of an amount of surfactant as follows: about or at least 0.1. Mu.l/ml, 0.2. Mu.l/ml, 0.4. Mu.l/ml, 0.6. Mu.l/ml, 0.8. Mu.l/ml, 1. Mu.l/ml, 1.5. Mu.l/ml, 2. Mu.l/ml, 2.5. Mu.l/ml, 5. Mu.l/ml, 10. Mu.l/ml, 25. Mu.l/ml or 50. Mu.l/ml. In certain embodiments, the enriched T cell composition is incubated in the presence of a surfactant at or about 2 μl/ml.
In some embodiments, the surfactant is or includes an agent that reduces the surface tension of a liquid and/or solid. For example, surfactants include fatty alcohols (e.g., sterols), polyoxyethylene glycol octylphenol ether (e.g., triton X-100), or polyoxyethylene glycol sorbitan alkyl esters (e.g., polysorbate 20, 40, 60). In certain embodiments, the surfactant is selected from polysorbate 80 (PS 80), polysorbate 20 (PS 20), poloxamer 188 (P188). In exemplary embodiments, the concentration of surfactant in the chemically defined feed medium is about 0.0025% to about 0.25% (v/v) PS80; about 0.0025% to about 0.25% (v/v) PS20; or about 0.1% to about 5.0% (w/v) P188.
In some embodiments, the surfactant is or includes an anionic surfactant, a cationic surfactant, a zwitterionic surfactant, or a nonionic surfactant added thereto. Suitable anionic surfactants include, but are not limited to, alkyl sulfonates, alkyl phosphates, alkyl phosphonates, potassium laurate, triethanolamine stearate, sodium lauryl sulfate, alkyl polyoxyethylene sulfates, sodium alginate, sodium dioctyl sulfosuccinate, phosphatidylglycerol, phosphatidylinosine, phosphatidylinositol, bis-phosphatidylglycerol, phosphatidylserine, phosphatidic acid and salts thereof, sodium carboxymethyl cellulose, cholic acid and other bile acids (e.g., cholic acid, deoxycholic acid, glycocholic acid, taurocholic acid, glycodeoxycholic acid) and salts thereof (e.g., sodium deoxycholate).
In some embodiments, suitable nonionic surfactants include: glycerol esters, polyoxyethylene fatty alcohol ethers, polyoxyethylene sorbitan fatty acid esters (polysorbate), polyoxyethylene fatty acid esters, sorbitan esters, glycerol monostearate, polyethylene glycol, polypropylene glycol, cetyl alcohol, cetylstearyl alcohol, stearyl alcohol, arylalkyl polyether alcohols, polyoxyethylene-polyoxypropylene copolymers (poloxamers), poloxamers, methyl cellulose, hydroxymethyl cellulose, hydroxypropyl methyl cellulose, amorphous cellulose, polysaccharides (including starch and starch derivatives, such as hydroxyethyl starch (HES)), polyvinyl alcohol, and polyvinylpyrrolidone. In certain embodiments, the nonionic surfactant is a polyoxyethylene and polyoxypropylene copolymer, and preferably is a block copolymer of propylene glycol and ethylene glycol. Such polymers are sold under the trade name POLOXAMER, sometimes also referred to as F68 or->P188. Polyoxyethylene fatty acid esters include those having a short alkyl chain. An example of such a surfactant is +.>HS15, polyethylene-660-hydroxystearate.
In some embodiments, suitable cationic surfactants may include, but are not limited to, natural phospholipids, synthetic phospholipids, quaternary ammonium compounds, benzalkonium chloride, cetyltrimethylammonium bromide, chitosan, lauryl dimethylbenzyl ammonium chloride, acyl carnitine hydrochloride, dimethyl Dioctadecyl Ammonium Bromide (DDAB), dioleoyl trimethylammonium propane (DOTAP), ditetradecyl trimethylammonium propane (dmtpap), dimethylaminoethane carbamoyl cholesterol (DC-Chol), 1, 2-diacylglycerol-3- (O-alkyl) phosphorylcholine, O-alkylphospholipid choline, alkylpyridinium halides, or long chain alkylamines (e.g., n-octylamine and oleamide).
Zwitterionic surfactants are electrically neutral, but have localized positive and negative charges within the same molecule. Suitable zwitterionic surfactants include, but are not limited to, zwitterionic phospholipids. Suitable phospholipids include phosphatidylcholine, phosphatidylethanolamine, diacyl-glycerol-phosphoethanolamine (e.g., ditetradecyl-glycerol-phosphoethanolamine (DMPE), dipalmitoyl-glycerol-phosphoethanolamine (DPPE), distearyl-glycerol-phosphoethanolamine (DSPE), and dioleoyl-glycerol-phosphoethanolamine (DOPE)). Mixtures of phospholipids (including anionic phospholipids and zwitterionic phospholipids) may be used in the present invention. Such mixtures include, but are not limited to, lysophospholipids, lecithins, or soybean phospholipids, or any combination thereof. Phospholipids (whether anionic, zwitterionic or mixtures of phospholipids) may be salted or desalted, hydrogenated or partially hydrogenated or be natural semisynthetic or synthetic.
In certain embodiments, the surfactant is a poloxamer, for example, poloxamer 188. In some embodiments, the T cell enriched composition is incubated in the presence of poloxamer in the following amounts: between 0.1 and 10.0. Mu.l/ml, between 0.2 and 2.5. Mu.l/ml, between 0.5 and 5. Mu.l/ml, between 1 and 3. Mu.l/ml or between 2 and 4. Mu.l/ml. In some embodiments, the T cell enriched composition is incubated in the presence of the following amounts of surfactants: about or at least 0.1. Mu.l/ml, 0.2. Mu.l/ml, 0.4. Mu.l/ml, 0.6. Mu.l/ml, 0.8. Mu.l/ml, 1. Mu.l/ml, 1.5. Mu.l/ml, 2. Mu.l/ml, 2.5. Mu.l/ml, 5. Mu.l/ml, 10. Mu.l/ml, 25. Mu.l/ml or 50. Mu.l/ml. In certain embodiments, the T cell enriched composition is incubated in the presence of poloxamer at or about 2 μl/ml.
In certain embodiments, the incubation is ended when the cells achieve a threshold amount, concentration, and/or expansion, such as by harvesting the cells. In particular embodiments, incubation ends when, for example, the cells achieve or achieve about or at least 1.5-fold amplification, 2-fold amplification, 2.5-fold amplification, 3-fold amplification, 3.5-fold amplification, 4-fold amplification, 4.5-fold amplification, 5-fold amplification, 6-fold amplification, 7-fold amplification, 8-fold amplification, 9-fold amplification, 10-fold amplification, or greater than 10-fold amplification with respect to and/or relative to the amount of cell density at the beginning or start of incubation. In some embodiments, the threshold expansion is, for example, about and/or relative to 4-fold expansion of the amount or density of cells at the beginning or initial incubation.
In some embodiments, incubation is ended when the cells achieve a threshold total amount of cells, e.g., a threshold cell count, such as by harvesting the cells. In some embodiments, incubation is terminated when the cells achieve a threshold Total Nucleated Cell (TNC) count. In some embodiments, incubation is ended when the cells achieve a threshold living cell amount (e.g., a threshold living cell count). In some embodiments, the threshold cell count is or is about or at least 50x10 6 Individual cells, 100x10 6 Individual cells, 200x10 6 Individual cells, 300x10 6 Individual cells, 400x10 6 Individual cells, 600x10 6 Individual cells, 800x10 6 Individual cells, 1000x10 6 Individual cells, 1200x10 6 Individual cells, 1400x10 6 Individual cells, 1600x10 6 Individual cells, 1800x10 6 Individual cells, 2000x10 6 Individual cells, 2500x10 6 Individual cells, 3000x10 6 Individual cells, 4000x10 6 Individual cells, 5000x10 6 Individual cells, 10,000x10 6 Individual cells, 12,000x10 6 Individual cells, 15,000x10 6 Individual cells or 20,000x10 6 Individual cells, or any of the preceding living cell thresholds. In certain embodiments, incubation is terminated when the cells achieve a threshold cell count. In some embodiments, the threshold cell count is achieved at about 6 hours, 12 hours, 24 hours, 36 hours, 1 day, 2 days, The incubation is ended within 3 days, 4 days, 5 days, 6 days or 7 days or more or within 6 hours, 12 hours, 24 hours, 36 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days or 7 days or more. In certain embodiments, incubation is terminated at or about 1 day after achieving a threshold cell count. In certain embodiments, the threshold density is, is about or at least 0.1x10 6 Individual cells/ml, 0.5x10 6 Individual cells/ml, 1X10 6 Individual cells/ml, 1.2x10 6 Individual cells/ml, 1.5x10 6 Individual cells/ml, 1.6x10 6 Individual cells/ml, 1.8x10 6 Individual cells/ml, 2.0x10 6 Individual cells/ml, 2.5x10 6 Individual cells/ml, 3.0x10 6 Individual cells/ml, 3.5x10 6 Individual cells/ml, 4.0x10 6 Individual cells/ml, 4.5x10 6 Individual cells/ml, 5.0x10 6 Individual cells/ml, 6X10 6 Individual cells/ml, 8x10 6 Individual cells/ml, or 10x10 6 Individual cells/ml, or any of the preceding living cell thresholds. In certain embodiments, incubation is terminated when the cells achieve a threshold density. In some embodiments, the incubation is ended at or about 6 hours, 12 hours, 24 hours, 36 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, or 7 days or more or within 6 hours, 12 hours, 24 hours, 36 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, or 7 days or more after achieving the threshold density. In certain embodiments, incubation is complete at or about 1 day after achieving the threshold density.
In some embodiments, the incubating step is performed for an amount of time required to achieve a threshold amount, density, and/or expansion for the cells. In some embodiments, incubation is performed for the following amount of time: is or is about or less than 6 hours, 12 hours, 18 hours, 24 hours, 36 hours, 48 hours, 60 hours, 72 hours, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 week, 2 weeks, 3 weeks, or 4 weeks. In particular embodiments, the average amount of time required to achieve a threshold density for cells of a plurality of separate compositions of isolated, enriched, and/or selected enriched T cells from different biological samples is about or less than 6 hours, 12 hours, 18 hours, 24 hours, 36 hours, 48 hours, 60 hours, 72 hours, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 week, 2 weeks, 3 weeks, or 4 weeks. In certain embodiments, the average amount of time required to achieve a threshold density for cells of a plurality of separate compositions of isolated, enriched, and/or selected enriched T cells from different biological samples is about or less than 6 hours, 12 hours, 18 hours, 24 hours, 36 hours, 48 hours, 60 hours, 72 hours, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 1 week, 2 weeks, 3 weeks, or 4 weeks.
In certain embodiments, the incubation step is performed for a minimum of 4, 5, 6, 7, 8, 9, or 10 days, and/or until the cells achieve the following threshold cell count (or number) or threshold viable cell count (or number) for 12, 24, 36, 1, 2, or 3 days: is or about 1000x10 6 Individual cells, 1200x10 6 Individual cells, 1400x10 6 Individual cells, 1600x10 6 Individual cells, 1800x10 6 Individual cells, 2000x10 6 Individual cells, 2500x10 6 Individual cells, 3000x10 6 Individual cells, 4000x10 6 Individual cells or 5000x10 6 Individual cells. In some embodiments, the culturing step is performed until the cells are achieved at or about 1200x10 6 Threshold cell count of individual cells and 1 day after a minimum of 10 days of culture, and/or until cells achieve at or about 5000x10 6 Threshold cell count for each cell 1 day. In some embodiments, the culturing step is performed until the cells are achieved at or about 1200x10 6 Threshold cell count of individual cells and 1 day after a minimum of 9 days of culture, and/or until cells achieve at or about 5000x10 6 Threshold cell count for each cell 1 day. In some embodiments, the culturing step is performed until the cells are achieved at or about 1000x10 6 Threshold cell count of individual cells and 1 day after a minimum of 8 days of culture, and/or until cells are achieved at or about 4000x10 6 Threshold cell count for each cell 1 day. In certain embodiments, the incubation is an expansion step and is performed for a minimum of 4, 5, 6, 7, 8, 9, or 10 days, and/or until the cells achieve a threshold cell count as follows12 hours, 24 hours, 36 hours, 1 day, 2 days, or 3 days after (or number of) or threshold viable cell count: is or about 1000x10 6 Individual cells, 1200x10 6 Individual cells, 1400x10 6 Individual cells, 1600x10 6 Individual cells, 1800x10 6 Individual cells, 2000x10 6 Individual cells, 2500x10 6 Individual cells, 3000x10 6 Individual cells, 4000x10 6 Individual cells or 5000x10 6 Individual cells. In some embodiments, the expansion step is performed until the cells achieve at or about 1200x10 6 Threshold cell count of individual cells and 1 day after a minimum of 10 days of expansion, and/or until cells achieve at or about 5000x10 6 Threshold cell count for each cell 1 day. In some embodiments, the expansion step is performed until the cells achieve at or about 1200x10 6 Threshold cell count of individual cells and 1 day after a minimum of 9 days of expansion, and/or until cells achieve at or about 5000x10 6 Threshold cell count for each cell 1 day. In some embodiments, the expansion step is performed until the cells are achieved at or about 1000x10 6 Threshold cell count of individual cells and 1 day after a minimum of 8 days of expansion, and/or until cells achieve at or about 4000x10 6 Threshold cell count for each cell 1 day. In some embodiments, the expansion step is performed until the cells achieve at or about 1400x10 6 Threshold cell count of individual cells and 1 day after a minimum of 5 days of expansion, and/or until cells achieve at or about 4000x10 6 Threshold cell count for each cell 1 day.
In some embodiments, incubating is performed for at least a minimum amount of time. In some embodiments, the incubation is performed for at least 14 days, at least 12 days, at least 10 days, at least 7 days, at least 6 days, at least 5 days, at least 4 days, at least 3 days, at least 2 days, at least 36 hours, at least 24 hours, at least 12 hours, or at least 6 hours, even if the threshold is achieved before the minimum amount of time. In some embodiments, in some cases, increasing the minimum amount of time to incubate may reduce activation and/or reduce the level of one or more activation markers in the incubated cells, the formulated cells, and/or the cells outputting the composition. In some embodiments, the minimum incubation time is counted from the determined point (e.g., the selection step; the thawing step; and/or the activation step) of the exemplary process to the day the cells were harvested.
In aspects of the provided embodiments, the density and/or concentration of cells or living cells is monitored during or performed during incubation, such as until a threshold amount, density and/or expansion as described is achieved. In some embodiments, such methods include those as described, including optical methods, including Digital Holographic Microscopy (DHM) or Differential Digital Holographic Microscopy (DDHM).
In certain embodiments, the cultured cells are export cells. In some embodiments, the composition of enriched T cells (e.g., engineered T cells) that have been incubated is an output composition of enriched T cells. In certain embodiments, the cd4+ T cells and/or cd8+ T cells that have been incubated are export cd4+ and/or cd8+ T cells. In certain embodiments, the composition enriched for cd4+ T cells (e.g., engineered cd4+ T cells) that has been incubated is an output composition enriched for cd4+ T cells. In some embodiments, the composition enriched for cd8+ T cells (e.g., engineered cd8+ T cells) that has been incubated is an output composition enriched for cd8+ T cells.
In some embodiments, the cells are incubated in the presence of one or more cytokines under conditions that promote proliferation and/or expansion. In certain embodiments, at least a portion of the incubation is performed with constant mixing and/or perfusion (e.g., mixing or perfusion controlled by a bioreactor). In some embodiments, the cells are incubated in the presence of one or more cytokines and with a surfactant (e.g., a poloxamer such as poloxamer 188) to reduce shear and/or shear stress from constant mixing and/or infusion. In some embodiments, the composition enriched for cd4+ T cells (e.g., engineered cd4+ T cells) is incubated in the presence of recombinant IL-2, IL-7, IL-15, and poloxamer, wherein at least a portion of the incubation is performed with constant mixing and/or perfusion. In certain embodiments, a composition enriched for cd8+ T cells (e.g., engineered cd8+ T cells) is incubated in the presence of recombinant IL-2, IL-15, and poloxamer, wherein at least a portion of the incubation is performed with constant mixing and/or perfusion. In some embodiments, the incubation is performed until the cells reach a threshold expansion of at least 4 times, for example, compared to when the incubation was initiated.
1. Monitoring cells during incubation
In some embodiments, the cells are monitored during the incubation step. Monitoring may be performed, for example, to determine (e.g., measure, quantify) cell morphology, cell viability, cell death, and/or cell concentration (e.g., living cell concentration). In some embodiments, the monitoring is performed manually, such as by a human operator. In some embodiments, the monitoring is performed by an automated system. Automated systems may require minimal or no manual input to monitor the cultured cells. In some embodiments, the monitoring is performed manually and by an automated system.
In certain embodiments, the cells are monitored by an automated system that does not require manual input. In some embodiments, the automated system is compatible with a bioreactor (e.g., a bioreactor described herein) such that cells undergoing incubation can be removed from the bioreactor, monitored, and then returned to the bioreactor. In some embodiments, monitoring and incubating occurs in a closed loop configuration. In some aspects, the automation system and the bioreactor remain sterile in a closed loop configuration. In embodiments, the automated system is sterile. In some embodiments, the automated system is an online system.
In some embodiments, the automated system comprises detecting cell morphology, cell viability, cell death, and/or cell concentration (e.g., viable cell concentration) using optical techniques (e.g., microscopy). Any optical technique suitable for determining, for example, cell characteristics, viability and concentration is contemplated herein. Non-limiting examples of useful optical techniques include bright field microscopy, fluorescence microscopy, differential Interference Contrast (DIC) microscopy, phase contrast microscopy, digital Holographic Microscopy (DHM), differential Digital Holographic Microscopy (DDHM), or combinations thereof. Differential digital holographic microscopy, DDHM and differential DHM may be used interchangeably herein. In certain embodiments, the automated system comprises a differential digital holographic microscope. In certain embodiments, the automated system comprises a differential digital holographic microscope, including an illumination device (e.g., laser, led). Descriptions of DDHM methods and uses can be found, for example, in the following documents: US 7,362,449; EP 1,631,788; US 9,904,248; and US 9,684,281, which is incorporated herein by reference in its entirety.
DDHM allows label-free, non-destructive imaging of cells, resulting in a high contrast holographic image. The image may be subject to segmentation and further analysis to obtain a plurality of morphological features that quantitatively describe the imaged subject (e.g., cultured cells, cell debris). In this way, various features (e.g., cell morphology, cell viability, cell concentration) can be estimated or calculated directly from the DDHM using steps such as image acquisition, image processing, image segmentation, and feature extraction. In some embodiments, the automated system includes a digital recording device to record the holographic image. In some embodiments, the automated system comprises a computer comprising an algorithm for analyzing the holographic image. In some embodiments, the automated system comprises a monitor and/or computer for displaying the results of the holographic image analysis. In some embodiments, the analysis is automated (i.e., can be performed without user input). Examples of suitable automated systems for monitoring cells during the incubation step include, but are not limited to, ovizio iLine F (Ovizio Imaging Systems NV/SA, brussell Belgium).
In certain embodiments, monitoring is performed continuously during the incubation step. In some embodiments, monitoring is performed in real time during the incubation step. In some embodiments, monitoring is performed at discrete points in time during the incubation step. In some embodiments, monitoring is performed at least once every 15 minutes for the duration of the incubation step. In some embodiments, monitoring is performed at least once every 30 minutes for the duration of the incubation step. In some embodiments, monitoring is performed at least once every 45 minutes for the duration of the incubation step. In some embodiments, the monitoring is performed at least once every hour for the duration of the incubation step. In some embodiments, monitoring is performed at least once every 2 hours for the duration of the incubation step. In some embodiments, monitoring is performed at least once every 4 hours for the duration of the incubation step. In some embodiments, monitoring is performed at least once every 6 hours for the duration of the incubation step. In some embodiments, monitoring is performed at least once every 8 hours for the duration of the incubation step. In some embodiments, monitoring is performed at least once every 10 hours for the duration of the incubation step. In some embodiments, monitoring is performed at least once every 12 hours for the duration of the incubation step. In some embodiments, monitoring is performed at least once every 14 hours for the duration of the incubation step. In some embodiments, monitoring is performed at least once every 16 hours for the duration of the incubation step. In some embodiments, monitoring is performed at least once every 18 hours for the duration of the incubation step. In some embodiments, monitoring is performed at least once every 20 hours for the duration of the incubation step. In some embodiments, monitoring is performed at least once every 22 hours for the duration of the incubation step. In some embodiments, the monitoring is performed at least once daily for the duration of the incubation step. In some embodiments, monitoring is performed at least once every two days for the duration of the incubation step. In some embodiments, monitoring is performed at least once every three days for the duration of the incubation step. In some embodiments, monitoring is performed at least once every four days for the duration of the incubation step. In some embodiments, monitoring is performed at least once every five days for the duration of the incubation step. In some embodiments, monitoring is performed at least once every six days for the duration of the incubation step. In some embodiments, monitoring is performed at least once every seven days for the duration of the incubation step. In some embodiments, monitoring is performed at least once every eight days for the duration of the incubation step. In some embodiments, monitoring is performed at least once every nine days for the duration of the incubation step. In some embodiments, monitoring is performed at least once every ten days for the duration of the incubation step. In some embodiments, the monitoring is performed at least once during the incubation step.
In some embodiments, the cellular characteristics that can be determined by monitoring (including using optical techniques such as DHM or DDHM) include cell viability, cell concentration, cell number, and/or cell density. In some embodiments, cell viability is characterized or determined. In some embodiments, cell concentration, density, and/or number are characterized or determined. In some embodiments, the viable cell concentration, viable cell number, and/or viable cell density is characterized or determined. In some embodiments, the cultured cells are monitored by an automated system until a threshold of expansion is reached as described above. In some embodiments, once the threshold for expansion is reached, the cultured cells are harvested, e.g., by automated or manual methods, e.g., by a human operator. The threshold for expansion may depend on the total concentration, density, and/or number of cultured cells as determined by the automated system. Alternatively, the threshold for expansion may depend on the concentration, density, and/or number of living cells.
In some embodiments, the harvested cells are formulated as described, e.g., in the presence of a pharmaceutically acceptable carrier. In some embodiments, the harvested cells are formulated in the presence of a cryoprotectant. In some embodiments, the efficacy of harvested cells of a therapeutic composition is assessed according to the methods provided in section I above. In some embodiments, the efficacy of the harvested cells of the therapeutic composition is assessed prior to cryopreservation. In some embodiments, the efficacy of the harvested cells of the therapeutic composition is assessed after cryopreservation.
E. Cell and therapeutic compositions for formulating recombinant receptor engineered cells
Also provided are compositions, including pharmaceutical compositions and formulations thereof, containing therapeutic cell compositions, the efficacy of which are evaluated according to the methods provided above (section I). In some embodiments, provided methods for making, generating or producing cell therapies and/or engineering cells can include formulating genetically engineered cells produced by the provided processing steps to produce a therapeutic cell composition comprising cells expressing a recombinant receptor. In some embodiments, provided methods related to the formulation of cells include processing transduced cells in a closed system, such as cells transduced and/or expanded using the processing steps described above. In some embodiments, the dose of cells comprising cells engineered with a recombinant antigen receptor (e.g., CAR or TCR) is provided as a composition or formulation, such as a pharmaceutical composition or formulation. Such compositions may be used according to the provided methods, such as for the prevention or treatment of diseases, conditions and disorders, or in detection, diagnosis and prognosis methods.
In some cases, processing the cells in one or more steps (e.g., performed in a centrifugal chamber and/or closed system) for making, generating, or producing cell therapies and/or engineering the cells may include formulating the cells before or after culturing (e.g., incubating and expanding) and/or one or more other processing steps as described, e.g., formulating genetically engineered cells produced by the provided transduction processing steps. In some cases, the cells may be formulated in an amount for dosage administration (e.g., for single unit dosage administration or multi-dosage administration). In some embodiments, the unit dose and/or dose administration is determined using the potency of the cells of the therapeutic composition determined according to the methods provided in section I above. In some embodiments, the efficacy of cells of a therapeutic composition is assessed according to the methods provided in section I for the purpose of determining a unit dose and/or dose administration. In some embodiments, provided methods related to the formulation of cells include processing transduced cells in a closed system, such as cells transduced and/or expanded using the processing steps described above.
In certain embodiments, one or more compositions of enriched T cells (e.g., engineered and cultured T cells, e.g., export T cells), therapeutic cell compositions, are formulated. In particular embodiments, one or more compositions enriched for T cells (e.g., engineered and cultured T cells, e.g., export T cells), therapeutic cell compositions, are formulated after the one or more compositions have been engineered and/or cultured. In certain embodiments, one or more of the compositions is an input composition. In some embodiments, one or more of the input compositions have been previously frozen and stored at low temperature and thawed prior to incubation.
In certain embodiments, the one or more therapeutic compositions enriched for T cells (e.g., engineered and cultured T cells, e.g., export T cells) are or comprise two separate compositions, e.g., separate engineered and/or cultured compositions, of enriched T cells. In particular embodiments, two separate therapeutic compositions enriched for T cells, e.g., two separate compositions enriched for cd4+ T cells and cd8+ T cells that are selected, isolated, and/or enriched, individually engineered, and individually cultured from the same biological sample, are formulated separately. In certain embodiments, the two separately therapeutic cell compositions include a composition enriched for cd4+ T cells, such as a composition engineered and/or incubated for cd4+ T cells. In particular embodiments, the two separately therapeutic cell compositions include compositions enriched for cd8+ T cells, such as compositions engineered and/or incubated for cd8+ T cells. In some embodiments, two separate therapeutic compositions enriched for cd4+ T cells and enriched for cd8+ T cells (e.g., a separate composition of engineered and cultured cd4+ T cells and engineered and cultured cd8+ T cells) are formulated separately. In some embodiments, the T cell enriched monotherapy composition is formulated. In certain embodiments, the monotherapy composition is a composition enriched for cd4+ T cells, such as a composition of engineered and/or incubated cd4+ T cells. In some embodiments, the monotherapy composition is a composition enriched for cd4+ and cd8+ T cells that have been combined from a separate composition prior to formulation.
In some embodiments, the separate therapeutic compositions enriched for cd4+ and cd8+ T cells (e.g., the separate compositions of engineered and incubated cd4+ and cd8+ T cells) are combined into a single therapeutic composition and formulated. In certain embodiments, the separately formulated therapeutic compositions enriched for cd4+ and enriched for cd8+ T cells are combined into a single therapeutic composition after the formulation has been performed and/or completed. In particular embodiments, the separate therapeutic compositions enriched for cd4+ and cd8+ T cells (e.g., the separate compositions of engineered and cultured cd4+ and cd8+ T cells) are formulated as separate compositions, respectively.
In some embodiments, the therapeutic composition of formulated enriched cd4+ T cells (e.g., engineered and cultured cd4+ T cells, e.g., exported cd4+ T cells) comprises at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or about 100% cd4+ T cells. In some embodiments, the composition comprises at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or at or about 100% of cd4+ T cells that express the recombinant receptor and/or have been transduced or transfected with the recombinant polynucleotide. In certain embodiments, the therapeutic composition formulated enriched for cd4+ T cells (e.g., engineered and cultured cd4+ T cells, e.g., exported cd4+ T cells) comprises less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1%, or less than 0.01% cd8+ T cells, and/or is free of cd8+ T cells, and/or is free or substantially free of cd8+ T cells.
In some embodiments, the therapeutic composition of formulated enriched cd8+ T cells (e.g., engineered and incubated cd8+ T cells, e.g., exported cd8+ T cells) comprises at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or about 100% cd8+ T cells. In certain embodiments, the therapeutic composition comprises at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, at least 99%, at least 99.5%, at least 99.9%, or at or about 100% of cd8+ T cells expressing the recombinant receptor and/or that have been transduced or transfected with the recombinant polynucleotide. In certain embodiments, a therapeutic composition enriched in cd8+ T cells (e.g., engineered and incubated cd8+ T cells, e.g., exported cd8+ T cells) incubated under stimulating conditions comprises less than 40%, less than 35%, less than 30%, less than 25%, less than 20%, less than 15%, less than 10%, less than 5%, less than 1%, less than 0.1%, or less than 0.01% cd4+ T cells, and/or is free of cd4+ T cells, and/or is free or substantially free of cd4+ T cells.
In certain embodiments, the formulated cell is an export cell. In some embodiments, the formulated therapeutic composition of enriched T cells (e.g., the formulated composition of engineered and cultured T cells) is an output composition of enriched T cells. In particular embodiments, the formulated cd4+ T cells and/or formulated cd8+ T cells are export cd4+ and/or cd8+ T cells. In particular embodiments, the formulated composition enriched for cd4+ T cells is an output composition enriched for cd4+ T cells. In some embodiments, the formulated composition enriched for cd8+ T cells is an output composition enriched for cd8+ T cells.
In some embodiments, the cells may be formulated into a container (e.g., a bag or vial). In some embodiments, the cells are formulated after the cells have achieved a threshold cell count, density, and/or between 0 and 10 days, between 0 and 5 days, between 2 and 7 days, between 0.5 and 4 days, or between 1 and 3 days after expansion during the incubation period. In certain embodiments, the cells are formulated at or about 12 hours, 18 hours, 24 hours, 1 day, 2 days, or 3 days or within 12 hours, 18 hours, 24 hours, 1 day, 2 days, or 3 days after threshold cell count, density, and/or expansion have been achieved during the incubation period. In some embodiments, the cells are formulated within or about 1 day after threshold cell counts, densities, and/or expansion have been achieved during incubation.
In some embodiments, the cells are formulated in a pharmaceutically acceptable buffer, which in some aspects may include a pharmaceutically acceptable carrier or excipient. In some embodiments, processing includes replacing the medium with a pharmaceutically acceptable or administration to the subject of the desired medium or formulation buffer. In some embodiments, the processing step may involve washing the transduced and/or expanded cells to replace the cells in a pharmaceutically acceptable buffer, which may include one or more optional pharmaceutically acceptable carriers or excipients. Examples of such pharmaceutical forms, including pharmaceutically acceptable carriers or excipients, may be any of the forms described below in connection with the administration of the cells and compositions to a subject. In some embodiments, the pharmaceutical composition contains cells in an amount effective to treat or prevent a disease or disorder (e.g., a therapeutically effective amount or a prophylactically effective amount).
The term "pharmaceutical formulation" refers to a formulation which is in a form such that the biological activity of the active ingredient contained therein is effective, and which is free of additional components having unacceptable toxicity to the subject to whom the formulation is administered.
By "pharmaceutically acceptable carrier" is meant an ingredient of the pharmaceutical formulation that is non-toxic to the subject in addition to the active ingredient. Pharmaceutically acceptable carriers include, but are not limited to, buffers, excipients, stabilizers, or preservatives.
In some aspects, the choice of vector depends in part on the particular cell and/or method of administration. Thus, there are a variety of suitable formulations. For example, the pharmaceutical composition may contain a preservative. Suitable preservatives may include, for example, methylparaben, propylparaben, sodium benzoate and benzalkonium chloride. In some aspects, a mixture of two or more preservatives is used. The preservative or mixture thereof is typically present in an amount of about 0.0001% to about 2% by weight of the total composition. Vectors are described, for example, in Remington's Pharmaceutical Sciences, 16 th edition, osol, a. Edit (1980). Pharmaceutically acceptable carriers are generally non-toxic to recipients at the dosages and concentrations employed, and include, but are not limited to: buffers such as phosphate, citrate and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (e.g., octadecyl dimethylbenzyl ammonium chloride, hexa methyl ammonium chloride, benzalkonium chloride, benzethonium chloride, phenol, butanol, or benzyl alcohol, alkyl parabens such as methyl or propyl parabens, catechol, resorcinol, cyclohexanol, 3-pentanol, and m-cresol); a low molecular weight (less than about 10 residues) polypeptide; proteins, such as serum albumin, gelatin or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars, such as sucrose, mannitol, trehalose, or sorbitol; salt-forming counterions, such as sodium; metal complexes (e.g., zinc-protein complexes); and/or nonionic surfactants such as polyethylene glycol (PEG).
In some aspects, a buffer is included in the composition. Suitable buffers include, for example, citric acid, sodium citrate, phosphoric acid, potassium phosphate, and various other acids and salts. In some aspects, a mixture of two or more buffers is used. The buffer or mixture thereof is typically present in an amount of about 0.001% to about 4% by weight of the total composition. Methods for preparing administrable pharmaceutical compositions are known. Exemplary methods are described in more detail in, for example, remington, the Science and Practice of Pharmacy, lippincott Williams & Wilkins; 21 st edition (month 1 of 2005 5).
In some embodiments, the pharmaceutical composition contains an amount (e.g., a therapeutically effective amount or a prophylactically effective amount) of the cells effective to treat or prevent the disease or disorder. In some embodiments, the treatment efficacy or prevention efficacy is monitored by periodic assessment of the subject being treated. For repeated administrations over several days or longer, depending on the condition, the treatment is repeated until the desired inhibition of disease symptoms occurs. However, other administration regimens may be useful and may be determined. The desired dose may be delivered by administering the composition by a single bolus, by administering the composition by multiple bolus injections, or by administering the composition by continuous infusion.
The cells may be administered using standard administration techniques, formulations and/or devices. Formulations and devices (e.g., syringes and vials) for storing and administering the compositions are provided. The administration of the cells may be autologous or heterologous. For example, immune response cells or progenitor cells can be obtained from one subject and administered to the same subject or a different compatible subject. The peripheral blood-derived immune response cells or their progeny (e.g., of in vivo, ex vivo, or in vitro origin) may be administered by local injection, including catheter administration, systemic injection, local injection, intravenous injection, or parenteral administration. When a therapeutic composition (e.g., a pharmaceutical composition containing genetically modified immune responsive cells) is administered, it is typically formulated in unit dose injectable form (solution, suspension, emulsion).
Formulations include those for oral, intravenous, intraperitoneal, subcutaneous, pulmonary, transdermal, intramuscular, intranasal, buccal, sublingual or suppository administration. In some embodiments, the cell population is administered parenterally. As used herein, the term "parenteral" includes intravenous, intramuscular, subcutaneous, rectal, vaginal and intraperitoneal administration. In some embodiments, the population of cells is administered to the subject using peripheral systemic delivery by intravenous, intraperitoneal, or subcutaneous injection.
In some embodiments, the compositions are provided as sterile liquid formulations, such as isotonic aqueous solutions, suspensions, emulsions, dispersions, or viscous compositions, which in some aspects may be buffered to a selected pH. Liquid formulations are generally easier to prepare than gels, other viscous compositions, and solid compositions. Additionally, the liquid composition is somewhat more convenient to administer, particularly by injection. On the other hand, the adhesive composition may be formulated within an appropriate viscosity range to provide longer contact times with specific tissues. The liquid or viscous composition may comprise a carrier, which may be a solvent or dispersion medium containing, for example, water, saline, phosphate buffered saline, polyols (e.g., glycerol, propylene glycol, liquid polyethylene glycol), and suitable mixtures thereof.
Sterile injectable solutions may be prepared by incorporating the cells in a solvent, for example, with suitable carriers, diluents or excipients such as sterile water, physiological saline, dextrose and the like. The composition may also be lyophilized. The compositions may contain auxiliary substances such as wetting, dispersing or emulsifying agents (e.g., methylcellulose), pH buffering agents, gelling or viscosity-enhancing additives, preservatives, flavoring agents, pigments and the like, depending on the route of administration and the desired formulation. In some aspects, standard text can be consulted to prepare a suitable formulation.
Various additives that enhance the stability and sterility of the composition may be added, including antimicrobial preservatives, antioxidants, chelating agents, and buffers. Prevention of the action of microorganisms can be ensured by different antibacterial and antifungal agents (e.g., parabens, chlorobutanol, phenol, ascorbic acid, and the like). Prolonged absorption of injectable pharmaceutical forms can be brought about by the use of agents which delay absorption (for example, aluminum monostearate and gelatin).
Formulations for in vivo administration are typically sterile. Sterility can be readily achieved, for example, by filtration through sterile filtration membranes.
In some embodiments, the formulation buffer contains a cryopreservative. In some embodiments, the cells are formulated with a cryopreservative solution containing 1.0% to 30% DMSO solution, such as 5% to 20% DMSO solution or 5% to 10% DMSO solution. In some embodiments, the cryopreservative solution is or contains, for example, PBS containing 20% DMSO and 8% Human Serum Albumin (HSA), or other suitable cell freezing medium. In some embodiments, the cryopreservative solution is or contains, for example, at least or about 7.5% DMSO. In some embodiments, the processing step may involve washing the transduced and/or expanded cells to replace the cells in the cryopreservative solution. In some embodiments, the cells are frozen, e.g., cryogenically frozen or cryogenically preserved, in a medium and/or solution having a final concentration of DMSO of or about 12.5%, 12.0%, 11.5%, 11.0%, 10.5%, 10.0%, 9.5%, 9.0%, 8.5%, 8.0%, 7.5%, 7.0%, 6.5%, 6.0%, 5.5%, or 5.0%, or DMSO of between 1% and 15%, between 6% and 12%, between 5% and 10%, or between 6% and 8%. In particular embodiments, the cells are frozen, e.g., cryogenically frozen or cryogenically preserved, in a medium and/or solution having a final concentration of HSA of or about 5.0%, 4.5%, 4.0%, 3.5%, 3.0%, 2.5%, 2.0%, 1.5%, 1.25%, 1.0%, 0.75%, 0.5% or 0.25%, or between 0.1% and 5%, between 0.25% and 4%, between 0.5% and 2%, or between 1% and 2%.
In particular embodiments, therapeutic compositions enriched for T cells (e.g., T cells that have been stimulated, engineered, and/or cultured) are formulated, cryogenically frozen, and then stored for a certain amount of time. In certain embodiments, the formulated cryogenically frozen cells are typically stored in multiple vials or containers until the cells are released for infusion. In some embodiments, vials or containers of a particular therapeutic composition may be used to perform the provided potency assay prior to infusion of the therapeutic cell composition. In particular embodiments, the formulated cryogenically frozen cells are stored for between 1 day and 6 months, between 1 month and 3 months, between 1 day and 14 days, between 1 day and 7 days, between 3 days and 6 days, between 6 months and 12 months, or longer than 12 months. In some embodiments, the cells are cryogenically frozen and stored for a duration of about, or for less than 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, or 7 days. In certain embodiments, after storage, the cells are thawed and administered to a subject. In certain embodiments, the cells are stored or stored for about 5 days.
In some embodiments, the cells are formulated in a pharmaceutically acceptable buffer (optionally including a cryoprotectant) having a volume of from 10mL to 1000mL or from about 10mL to about 1000mL, such as at least or about 50mL, 100mL, 200mL, 300mL, 400mL, 500mL, 600mL, 700mL, 800mL, 900mL or 1000mL. In some embodiments, the therapeutic cell composition is stored in a container (e.g., one or more vials or bags). In some embodiments, the vial or bag generally contains the cells to be administered, e.g., one or more unit doses thereof. The unit dose may be the amount or number of cells to be administered to the subject, or twice the number (or more) of cells to be administered. It may be the lowest dose or the lowest possible dose of cells to be administered to the subject.
In some embodiments, each container (e.g., bag or vial) individually contains a unit dose of cells. Thus, in some embodiments, each container contains the same or substantially the same number of cells. In some embodiments, each unit dose contains at least or about at least 1x10 6 、2x10 6 、5x10 6 、1x10 7 、5x10 7 Or 1x10 8 Individual engineered cells, total cells, T cells or PBMCs. In some embodiments, the volume of the formulated cell composition in each container (e.g., bag or vial) is 10mL to 100mL, such as at least or about 20mL, 30mL, 40mL, 50mL, 60mL, 70mL, 80mL, 90mL, or 100mL. In some embodiments, the cells in the container (e.g., bag or vial) may be cryopreserved. In some embodiments, the container (e.g., vial) may be stored in liquid nitrogen until further use.
In some embodiments, the potency (e.g., relative potency) of a cell of a composition comprising a recombinant receptor-expressing cell (e.g., CAR-expressing cell) as produced by a method comprising one or more of the steps is determined or measured using the potency assays described herein. In some embodiments, cells (e.g., CAR expressing cells) of a composition comprising recombinant receptor expressing cells (e.g., CAR expressing cells) can be administered to a subject (e.g., produced by a method comprising one or more of the steps and/or whose efficacy has been determined) to treat a disease or disorder.
III recombinant receptors
In some embodiments, the provided methods for determining the efficacy (e.g., relative potency) of a therapeutic cell composition are performed or carried out on cells from a composition that contains or expresses or is engineered to contain or express a recombinant receptor, e.g., a Chimeric Antigen Receptor (CAR) or a T Cell Receptor (TCR). In certain embodiments, the methods provided herein produce and/or are capable of producing cells or populations or compositions containing and/or enriched for cells engineered to express or contain recombinant proteins and the efficacy of such produced engineered cells can be determined or measured. In various embodiments, provided methods are performed on a cellular composition, such as an immune cell (e.g., a T cell), which is a cell engineered, transformed, transduced or transfected to express one or more recombinant receptors.
In some embodiments, provided methods are used to assess the efficacy of an engineered cell, such as an immune cell, such as a T cell, that expresses one or more recombinant receptors. Receptors include antigen receptors and receptors containing one or more components thereof. Recombinant receptors can include chimeric receptors (such as those containing a ligand binding domain or binding fragment thereof and an intracellular signaling domain or region), functional non-TCR antigen receptors, chimeric Antigen Receptors (CARs), T Cell Receptors (TCRs) (such as recombinant or transgenic TCRs), chimeric autoantibody receptors (CAARs), and components of any of the foregoing. Recombinant receptors, such as CARs, typically include an extracellular antigen (or ligand) binding domain linked (in some aspects by a linker and/or one or more transmembrane domains) to one or more intracellular signaling components. In some embodiments, the engineered cells express two or more receptors containing different components, domains, or regions. In some aspects, two or more receptors allow for spatial or temporal modulation or control of the specificity, activity, antigen (or ligand) binding, function, and/or expression of recombinant receptors.
A. Chimeric Antigen Receptor (CAR)
In some embodiments of the provided methods, a chimeric receptor (e.g., chimeric antigen receptor) contains one or more domains that combine a ligand binding domain (e.g., an antibody or antibody fragment) that provides specificity for a desired target (e.g., an antigen (e.g., a tumor antigen)) with an intracellular signaling domain. In some embodiments, the intracellular signaling domain is an activating intracellular domain portion, such as a T cell activating domain, thereby providing a primary activation signal. In some embodiments, the intracellular signaling domain contains or otherwise contains a costimulatory signaling domain to facilitate effector function. In some embodiments, the chimeric receptor, when genetically engineered into immune cells, can modulate T cell activity, and in some cases can modulate T cell differentiation or homeostasis, thereby producing genetically engineered cells with improved longevity, survival, and/or persistence in vivo, such as for adoptive cell therapy methods.
Exemplary antigen receptors (including CARs) and methods of engineering and introducing such receptors into cells include, for example, those described in the following documents: international patent application publication nos. WO 200014257, WO 2013126726, WO 2012/129514, WO 2014031687, WO 2013/166321, WO 2013/071154, WO 2013/123061, WO 2015/168413, WO 2016/030414; U.S. patent application publication nos. US2002131960, US2013287748, US20130149337, US20190389925; U.S. patent No.: 6,451,995, 7,446,190, 8,252,592, 8,339,645, 8,398,282, 7,446,179, 6,410,319, 7,070,995, 7,265,209, 7,354,762, 7,446,191, 8,324,353, 8,479,118, 10,266,580; european patent application number EP 2537416; and/or those described in the following documents: sadelain et al, cancer discover.2013, month 4; 3 (4) 388-398; davila et al (2013) PLoS ONE 8 (4): e61338; turtle et al, curr. Opin. Immunol, 10, 2012; 24 633-39; wu et al, cancer,2012, 3, 18 (2): 160-75. In some aspects, antigen receptors include CARs as described in U.S. Pat. No. 7,446,190, and those described in international patent application publication No. WO/2014055668 A1. Examples of CARs include CARs as disclosed in any of the above publications, e.g., WO 2014031687, US 8,339,645, US 7,446,179, US2013/0149337, U.S. Pat. nos. 7,446,190, U.S. Pat. No. 8,389,282; kochenderfer et al 2013,Nature Reviews Clinical Oncology,10,267-276 (2013); wang et al (2012) J.Immunother35 (9): 689-701; and Brentjens et al, sci Transl Med.2013 (177). See also WO 2014031687, US 8,339,645, US 7,446,179, US2013/0149337, U.S. Pat. No. 7,446,190 and U.S. Pat. No. 8,389,282.
Chimeric receptors, such as CARs, typically include an extracellular target binding domain (e.g., an antigen binding domain), such as, for example, a portion of an antibody molecule, typically a Variable Heavy (VH) chain region and/or a Variable Light (VL) chain region of an antibody, such as a scFv antibody fragment.
In some embodiments, the antigen to which the receptor is targeted is a polypeptide. In some embodiments, it is a carbohydrate or other molecule. In some embodiments, the antigen is selectively expressed or over-expressed on cells of a disease or disorder (e.g., tumor or pathogenic cells) as compared to normal or non-targeted cells or tissues. In other embodiments, the antigen is expressed on normal cells and/or on engineered cells.
In some embodiments, the antigen is or comprises αvβ6 integrin (avb 6 integrin), B Cell Maturation Antigen (BCMA), B7-H3, B7-H6, carbonic anhydrase 9 (CA 9, also known as CAIX or G250), cancer-testis antigen, cancer/testis antigen 1B (CTAG, also known as NY-ESO-1 and rage-2), carcinoembryonic antigen (CEA), cyclin A2, C-C motif chemokine ligand 1 (CCL-1), CD19, CD20, CD22, CD23, CD24, CD30, CD33, CD38, CD44v6, CD44v7/8, CD123, CD133, CD138, CD171, chondroitin sulfate proteoglycan 4 (CSPG 4), epidermal growth factor protein (EGFR), epidermal growth factor receptor type III mutant (EGFR vIII), epithelial glycoprotein 2 (EPG-2), epithelial glycoprotein 40 (EPG-40), hepadin B2, liver hormone receptor A2 (fcfc receptor 5), and the like receptor 5; also known as Fc receptor homolog 5 or FCRH 5), fetal acetylcholine receptor (fetal AchR), folic acid binding protein (FBP), folic acid receptor alpha, ganglioside GD2, O-acetylated GD2 (OGD 2), ganglioside GD3, glycoprotein 100 (gp 100), glypican-3 (GPC 3), G-protein coupled receptor 5D (GPRC 5D), her2/neu (receptor tyrosine kinase erb-B2), her3 (erb-B3), her4 (erb-B4), erbB dimer, human high molecular weight melanomA-Associated antigen (HMW-MAA), hepatitis B surface antigen, human leukocyte antigen A1 (HLa-A1), human leukocyte antigen A2 (HLa-A2), IL-22 receptor alpha (IL-22 ra), IL-13 receptor alpha 2 (IL-13 ra 2), kinase insert domain receptor (kdr), kappa light chain, L1 cell adhesion molecule (L1-CAM), CE7 epitope of L1-CAM, protein 8 family member a containing leucine rich repeat (LRRC 8A), lewis Y, melanomA-Associated antigen (MAGE) -A1, MAGE-A3, MAGE-A6, MAGE-a10, mesothelin (MSLN), c-Met, murine cytomegalovirus (MUC 1), MUC16, natural killer cell group 2 member D (g 2D) ligand, melanoma (MART 1), human cell adhesion molecule (nct 1), human prostate specific antigen (TRP), protein-associated protein (TRP 1), protein-specific receptor (TRP 1, p 2), protein-associated tumor antigen (TRP 1, p-specific tumor antigen (TRP 1), protein receptor-associated protein 2 (TRP 1), tumor-associated tumor antigen (TRP 1, tumor antigen (tumor antigen-specific tumor antigen), projection 1, tumor antigen (TRP 1, tumor antigen (tumor antigen), projection 1, tumor antigen (tumor antigen), tumor antigen (tumor antigen) protein 2), also known as dopachrome tautomerase, dopachrome delta isomerase, or DCT), vascular Endothelial Growth Factor Receptor (VEGFR), vascular endothelial growth factor receptor 2 (VEGFR 2), nephroblastoma 1 (WT-1), pathogen-specific or pathogen-expressed antigen, or an antigen comprising or associated with a universal tag, and/or a biotinylated molecule, and/or a molecule expressed by HIV, HCV, HBV or other pathogens. In some embodiments, the receptor-targeted antigen comprises an antigen associated with a B cell malignancy, such as any of a number of known B cell markers. In some embodiments, the antigen is or comprises CD20, CD19, CD22, ROR1, CD45, CD21, CD5, CD33, igκ, igλ, CD79a, CD79b, or CD30. In some embodiments, the antigen is or includes an antigen that is characteristic of or expressed by a pathogen. In some embodiments, the antigen is a viral antigen (e.g., a viral antigen from HIV, HCV, HBV, etc.), a bacterial antigen, and/or a parasitic antigen.
In some embodiments, the antibody is an antigen-binding fragment (e.g., scFv) comprising one or more linkers that connect two antibody domains or regions (e.g., a heavy chain Variable (VH) region and a light chain Variable (VL) region). The linker is typically a peptide linker, e.g., a flexible and/or soluble peptide linker. Linkers include those that are rich in glycine and serine and/or, in some cases, threonine. In some embodiments, the linker further comprises charged residues, such as lysine and/or glutamic acid, which may improve solubility. In some embodiments, the linker further comprises one or more prolines. In some aspects, a glycine and serine (and/or threonine) rich linker comprises at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% of one or more such amino acids. In some embodiments, they comprise at least or about 50%, 55%, 60%, 70% or 75% glycine, serine and/or threonine. In some embodiments, the linker consists essentially entirely of glycine, serine, and/or threonine. The length of the linker is typically between about 5 and about 50 amino acids, typically between or about 10 and or about 30, e.g., 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30, and in some examples between 10 and 25 amino acids in length. Exemplary linkers include linkers having a different number of repeats of sequences GGGGS (4 GS; SEQ ID NO: 22) or GGGS (3 GS; SEQ ID NO: 23), such as between 2, 3, 4 and 5 repeats of such sequences. Exemplary linkers include those having or consisting of the sequences set forth in SEQ ID NO. 24 (GGGGSGGGGSGGGGS), SEQ ID NO. 25 (GSTSGSGKPGSGEGSTKG), or SEQ ID NO. 26 (SRGGGGSGGGGSGGGGSLEMA).
In some embodiments, the receptor-targeted antigen comprises an antigen associated with a B cell malignancy, such as any of a number of known B cell markers. In some embodiments, the receptor-targeted antigen is CD20, CD19, CD22, ROR1, CD45, CD21, CD5, CD33, igκ, igλ, CD79a, CD79b, or CD30.
In some embodiments, the antigen or antigen binding domain is CD19. In some embodiments, the scFv comprises a VH and a VL derived from an antibody or antibody fragment specific for CD19. In some embodiments, the antibody or antibody fragment that binds CD19 is a mouse-derived antibody, such as FMC63 and SJ25C1. In some embodiments, the antibody or antibody fragment is a human antibody, e.g., as described in U.S. patent publication No. US 2016/0152723.
The term "antibody" is used herein in its broadest sense and includes polyclonal and monoclonal antibodies, including whole antibodies and functional (antigen-binding) antibody fragments, including fragment antigen-binding (Fab) fragments, F (ab ') 2 fragments, fab' fragments, fv fragments, recombinant IgG (IgG) fragments, heavy chain Variable (VH) regions capable of specifically binding an antigen, single chain antibody fragments (including single chain variable fragments (scFv)), and single domain antibodies (e.g., sdAb, sdFv, nanobody) fragments. The term encompasses genetically engineered and/or otherwise modified forms of immunoglobulins, such as intracellular antibodies, peptibodies, chimeric antibodies, fully human antibodies, humanized antibodies and heteroconjugated antibodies, multispecific (e.g., bispecific or trispecific) antibodies, diabodies, triabodies and tetrabodies, tandem diabodies, tandem triabodies. Unless otherwise indicated, the term "antibody" should be understood to encompass functional antibody fragments thereof, also referred to herein as "antigen-binding fragments". The term also encompasses whole or full length antibodies, including antibodies of any class or subclass (including IgG and subclasses thereof, igM, igE, igA and IgD).
The terms "complementarity determining region" and "CDR" are synonymous with "hypervariable region" or "HVR," and are known in the art to refer to non-contiguous amino acid sequences within the variable region of an antibody that confer antigen specificity and/or binding affinity. Typically, there are three CDRs (CDR-H1, CDR-H2, CDR-H3) in each heavy chain variable region, and three CDRs (CDR-L1, CDR-L2, CDR-L3) in each light chain variable region. "framework region" and "FR" are known in the art to refer to the non-CDR portions of the variable regions of the heavy and light chains. Typically, there are four FRs (FR-H1, FR-H2, FR-H3 and FR-H4) in each full-length heavy chain variable region, and four FRs (FR-L1, FR-L2, FR-L3 and FR-L4) in each full-length light chain variable region.
The exact amino acid sequence boundaries for a given CDR or FR can be readily determined using any of a number of well known schemes, including those described in the following documents: kabat et al (1991), "Sequences of Proteins of Immunological Interest," 5 th edition Public Health Service, national Institutes of Health, bethesda, MD ("Kabat" numbering scheme); al-Lazikani et Al, (1997) JMB 273,927-948 ("Chothia" numbering scheme); macCallum et al, J.mol. Biol.262:732-745 (1996), "anti-body-antigen interactions: contact analysis and binding site topography," J.mol. Biol.262,732-745 "(" Contact "numbering scheme); lefranc MP et al, "IMGT unique numbering for immunoglobulin and T cell receptor variabledomains and Ig superfamily V-like domains," Dev Comp Immunol, month 1 2003; 27 (1) 55-77 ("IMGT" numbering scheme); honyger A and Pluckthun A, "Yet another numbering scheme for immunoglobulin variabledomains: an automatic modeling and analysis tool," J Mol Biol, 6/8/2001; 309 (3) 657-70 ("Aho" numbering scheme); and Martin et al, "Modeling antibody hypervariable loops: a combined algorithm," PNAS,1989,86 (23): 9268-9272 ("AbM" numbering scheme).
The boundaries of a given CDR or FR may vary depending on the scheme used for identification. For example, the Kabat scheme is based on structural alignment, while the Chothia scheme is based on structural information. Numbering of the Kabat and Chothia protocols is based on the most common antibody region sequence length, with insertions provided by insert letters such as "30a" and deletions in some antibodies. Both of these schemes place certain insertions and deletions ("indels") at different positions, resulting in different numbers. The Contact scheme is based on analysis of complex crystal structures and is similar in many respects to the Chothia numbering scheme. The AbM protocol is a compromise between Kabat and Chothia definitions and is a protocol based on the use of Oxford Molecular's AbM antibody modeling software.
Table 1 lists exemplary location boundaries for CDR-L1, CDR-L2, CDR-L3 and CDR-H1, CDR-H2, CDR-H3 identified by the Kabat, chothia, abM and Contact schemes, respectively. For CDR-H1, residue numbers are listed using the two numbering schemes of Kabat and Chothia. FR is located between the CDRs, e.g., FR-L1 is located before CDR-L1, FR-L2 is located between CDR-L1 and CDR-L2, FR-L3 is located between CDR-L2 and CDR-L3, etc. It should be noted that because the Kabat numbering scheme shown places insertions at H35A and H35B, when numbered using the Kabat numbering convention shown, the ends of the Chothia CDR-H1 loop vary between H32 and H34 depending on the length of the loop.
Thus, unless otherwise specified, noIt is to be understood that a "CDR" or "complementarity determining region" or a separately designated CDR (e.g., CDR-H1, CDR-H2, CDR-H3) of a given antibody or region thereof (e.g., variable region thereof) encompasses one (or a particular) complementarity determining region as defined by any of the foregoing schemes or other known schemes. For example, in stating that a particular CDR (e.g., CDR-H3) contains a given V H Or V L In the case of the amino acid sequence of a corresponding CDR in the region amino acid sequence, it is to be understood that such CDR has the sequence of the corresponding CDR (e.g., CDR-H3) within the variable region, as defined by any of the foregoing schemes or other known schemes. In some embodiments, specific CDR sequences are specified. Exemplary CDR sequences of the provided antibodies are described using various numbering schemes, but it should be understood that the provided antibodies may include CDRs as described according to any other of the above-described numbering schemes or other numbering schemes known to the skilled artisan.
Likewise, unless otherwise specified, the FR of a given antibody or region thereof, such as its variable region, or a separately specified FR(s) (e.g., FR-H1, FR-H2, FR-H3, FR-H4) should be understood to encompass one (or a particular) framework region as defined by any known scheme. In some cases, an identification scheme for identifying a particular CDR, FR, or a plurality of particular FR or CDRs is specified, such as a CDR defined by Kabat, chothia, abM or Contact methods or other known schemes. In other cases, specific amino acid sequences of CDRs or FR are given.
The term "variable region" or "variable domain" refers to a domain of an antibody that is involved in the binding of the antibody to an antigen in the heavy or light chain of the antibody. The variable regions of the heavy and light chains of natural antibodies (V respectively H And V L ) Typically have a similar structure, wherein each domain comprises four conserved Framework Regions (FR) and three CDRs. (see, e.g., kit et al Kuby Immunology, 6 th edition, w.h. freeman and co., p 91 (2007). Singular V) H Or V L The domain may be sufficient to confer antigen binding specificity. In addition, V from antigen-binding antibodies can be used H Or V L Domain isolation of antibodies binding to specific antigens for separate selection of complementary V L Or V H Library of domains. Ginseng radixSee, for example, portolano et al, J.Immunol.150:880-887 (1993); clarkson et al Nature 352:624-628 (1991).
Antibodies included in the provided CARs include antibody fragments. An "antibody fragment" or "antigen-binding fragment" refers to a molecule other than an intact antibody that comprises a portion of the intact antibody that binds to an antigen to which the intact antibody binds. Examples of antibody fragments include, but are not limited to Fv, fab, fab ', fab ' -SH, F (ab ') 2 The method comprises the steps of carrying out a first treatment on the surface of the A diabody; a linear antibody; heavy chain variable (V H ) Regions, single chain antibody molecules (e.g., scFv) and containing only V H A single domain antibody of the region; and multispecific antibodies formed from antibody fragments. In some embodiments, the antigen binding domain in a provided CAR is or includes a polypeptide comprising a variable heavy chain (V H ) Region and variable light chain (V L ) Antibody fragments of the region. In particular embodiments, the antibody is a heavy chain variable (V H ) Region and/or light chain variable (V L ) Single chain antibody fragments (e.g., scFv) of the region.
In some embodiments, the scFv is derived from FMC63.FMC63 is typically a mouse monoclonal IgG1 antibody raised against human-derived Nalm-1 and Nalm-16 expressing CD19 cells (Ling, N.R. et al (1987) Leucocyte typing III.302). In some embodiments, the FMC63 antibody comprises CDRH1 and H2 as set forth in SEQ ID NO. 27 and 28, respectively, and CDRH3 as set forth in SEQ ID NO. 29 or 30, and CDRL1 as set forth in SEQ ID NO. 55, and CDR L2 as set forth in SEQ ID NO. 32 or 33, and CDR L3 as set forth in SEQ ID NO. 34 or 35. In some embodiments, the FMC63 antibody comprises a heavy chain variable region (V H ) And a light chain variable region (V) comprising the amino acid sequence of SEQ ID NO. 37 L )。
In some embodiments, the scFv comprises a variable light chain comprising the CDRL1 sequence of SEQ ID NO. 31, the CDRL2 sequence of SEQ ID NO. 32, and the CDRL3 sequence of SEQ ID NO. 34, and/or a variable heavy chain comprising the CDRH1 sequence of SEQ ID NO. 27, the CDRH2 sequence of SEQ ID NO. 28, and the CDRH3 sequence of SEQ ID NO. 29. In some embodiments, the scFv comprises a variable heavy chain region as set forth in SEQ ID NO:36 and a variable light chain region as set forth in SEQ ID NO: 37. In some implementations In embodiments, the variable heavy chain and the variable light chain are linked by a linker. In some embodiments, the linker is as shown in SEQ ID NO. 25. In some embodiments, the scFv comprises V in turn H Linker and V L . In some embodiments, the scFv comprises V in turn L Linker and V H . In some embodiments, the scFv is encoded by the nucleotide sequence shown in SEQ ID NO:38 or a sequence that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 38. In some embodiments, the scFv comprises the amino acid sequence shown in SEQ ID NO:39 or a sequence exhibiting at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO: 39.
In some embodiments, the scFv is derived from SJ25C1.SJ25C1 is a mouse monoclonal IgG1 antibody raised against human-derived Nalm-1 and Nalm-16 expressing CD19 (Ling, N.R. et al (1987) Leucocyte typing III.302). In some embodiments, the SJ25C1 antibody comprises the CDRH1, H2, and H3 sequences shown in SEQ ID NOS: 51-53, respectively, and CDRL1, L2, and L3 sequences shown in SEQ ID NOS: 48-50, respectively. In some embodiments, the SJ25C1 antibody comprises a heavy chain variable region (V H ) And a light chain variable region (V) comprising the amino acid sequence of SEQ ID NO. 47 L )。
In some embodiments, the scFv comprises a variable light chain comprising the CDRL1 sequence of SEQ ID NO. 48, the CDRL2 sequence of SEQ ID NO. 49, and the CDRL3 sequence of SEQ ID NO. 50, and/or a variable heavy chain comprising the CDRH1 sequence of SEQ ID NO. 51, the CDRH2 sequence of SEQ ID NO. 52, and the CDRH3 sequence of SEQ ID NO. 53. In some embodiments, the scFv comprises a variable heavy chain region as set forth in SEQ ID NO. 46 and a variable light chain region as set forth in SEQ ID NO. 47. In some embodiments, the variable heavy chain and the variable light chain are linked by a linker. In some embodiments, the linker is as shown in SEQ ID NO. 24. In some embodiments, the scFv comprises V in order H Linker and V L . In some embodiments, the scFv are packaged in sequenceContaining V L Linker and V H . In some embodiments, the scFv comprises the amino acid sequence shown in SEQ ID NO. 54 or a sequence exhibiting at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity to SEQ ID NO. 54.
In some embodiments, the antibody or antigen binding fragment (e.g., scFv or V H Domain) specifically recognizes an antigen (e.g., BCMA). In some embodiments, the antibody or antigen binding fragment is derived from, or is a variant of, an antibody or antigen binding fragment that specifically binds BCMA.
In some embodiments, the CAR is an anti-BCMA CAR that is specific for BCMA (e.g., human BCMA). Chimeric antigen receptors containing anti-BCMA antibodies (including mouse anti-human BCMA antibodies and human anti-human antibodies) and cells expressing such chimeric receptors have been previously described. See Carpenter et al, clin Cancer Res.,2013,19 (8): 2048-2060, WO 2016/090320,WO 2016090327,WO 2010104949A2 and WO 2017173256. In some embodiments, the antigen or antigen binding domain is BCMA. In some embodiments, the scFv comprises a VH and a VL derived from an antibody or antibody fragment specific for BCMA. In some embodiments, antibodies or antibody fragments that bind BCMA are or contain VH and VL from antibodies or antibody fragments described in international patent application publication nos. WO 2016/090327 and WO 2016/090320.
In some embodiments, the anti-BCMACAR comprises an antigen binding domain (e.g., scFv) comprising a variable heavy chain (V H ) Region and/or variable light chain (V L ) A zone. In some embodiments, the antigen binding domain (e.g., scFv) comprises a V set forth in SEQ ID NO:55 H And V shown in SEQ ID NO. 56 L . In some embodiments, the antigen binding domain (e.g., scFv) comprises a V set forth in SEQ ID NO:57 H And V shown in SEQ ID NO 58 L . In some embodiments, the antigen binding domain (e.g., scFv) comprises a V set forth in SEQ ID NO:59 H And SEQ ID NO. 60V L . In some embodiments, the antigen binding domain (e.g., scFv) comprises a V set forth in SEQ ID NO:61 H And V shown in SEQ ID NO. 62 L . In some embodiments, the antigen binding domain (e.g., scFv) comprises a V set forth in SEQ ID NO. 63 H And V shown in SEQ ID NO. 64 L . In some embodiments, the antigen binding domain (e.g., scFv) comprises a V set forth in SEQ ID NO:65 H And V shown in SEQ ID NO 66 L . In some embodiments, the antigen binding domain (e.g., scFv) comprises a V set forth in SEQ ID NO:67 H And V shown in SEQ ID NO. 68 L . In some embodiments, V H Or V L With a V exhibiting a value similar to any of the foregoing H Or V L At least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity and retains amino acid sequence binding to BCMA. In some embodiments, V H The region being located at V L The amino terminus of the region. In some embodiments, V H The region being located at V L The carboxy terminus of the region.
In some embodiments, the antigen or antigen binding domain is GPRC5D. In some embodiments, the scFv comprises a VH and a VL derived from an antibody or antibody fragment specific for GPRC5D. In some embodiments, the antibodies or antibody fragments that bind GPRC5D are or contain VH and VL from antibodies or antibody fragments described in International patent application publication Nos. WO 2016/090329 and WO 2016/090312.
In some aspects, the CAR contains a ligand (e.g., antigen) binding domain that binds or recognizes (e.g., specifically binds) a universal tag or universal epitope. In some aspects, the binding domain may bind a molecule, tag, polypeptide, and/or epitope, which may be linked to a different binding molecule (e.g., an antibody or antigen binding fragment) that recognizes an antigen associated with a disease or disorder. Exemplary tags or epitopes include dyes (e.g., fluorescein isothiocyanate) or biotin. In some aspects, a binding molecule (e.g., an antibody or antigen binding fragment) is linked to a tag that recognizes an antigen (e.g., a tumor antigen) associated with a disease or disorder, and an engineered cell expresses a CAR specific for the tag to effect cytotoxicity or other effector function of the engineered cell. In some aspects, the specificity of the CAR for an antigen associated with a disease or disorder is provided by a tagged binding molecule (e.g., an antibody), and different tagged binding molecules can be used to target different antigens. Exemplary CARs specific for a universal tag or universal epitope include, for example, those described in the following documents: U.S.9,233,125; WO 2016/030414; urbanska et al, (2012) Cancer Res 72:1844-1852; and Tamada et al, (2012) Clin Cancer Res 18:6436-6445.
In some embodiments, the antigen is or includes an antigen that is characteristic of or expressed by a pathogen. In some embodiments, the antigen is a viral antigen (e.g., a viral antigen from HIV, HCV, HBV, etc.), a bacterial antigen, and/or a parasitic antigen. In some embodiments, the CAR contains a TCR-like antibody, such as an antibody or antigen-binding fragment (e.g., scFv), that specifically recognizes an intracellular antigen (e.g., a tumor-associated antigen) that is present on the cell surface as a Major Histocompatibility Complex (MHC) -peptide complex. In some embodiments, antibodies or antigen binding portions thereof that recognize MHC-peptide complexes may be expressed on cells as part of a recombinant receptor (e.g., antigen receptor). Antigen receptors include functional non-T Cell Receptor (TCR) antigen receptors, such as Chimeric Antigen Receptors (CARs). In some embodiments, a CAR containing an antibody or antigen binding fragment that exhibits TCR-like specificity for a peptide-MHC complex may also be referred to as a TCR-like CAR. In some embodiments, the CAR is a TCR-like CAR and the antigen is a processed peptide antigen, such as a peptide antigen of an intracellular protein, that is recognized on the cell surface in the context of an MHC molecule as the TCR. In some embodiments, in some aspects, the extracellular antigen-binding domain specific for the MHC-peptide complex of a TCR-like CAR is linked to one or more intracellular signaling components by a linker and/or one or more transmembrane domains. In some embodiments, such molecules may generally mimic or resemble signals through natural antigen receptors (e.g., TCRs), as well as signals through combinations of such receptors with co-stimulatory receptors.
Reference to "major histocompatibility complex" (MHC) refers to a protein, typically a glycoprotein, containing polymorphic peptide binding sites or grooves, which in some cases may be complexed with peptide antigens of a polypeptide, including peptide antigens processed by cellular machinery. In some cases, MHC molecules may be displayed or expressed on the cell surface, including as complexes with peptides, i.e., MHC-peptide complexes, for presenting antigens having a conformation recognizable by antigen receptors on T cells (e.g., TCR or TCR-like antibodies). Generally, MHC class I molecules are heterodimers with a transmembrane alpha chain (in some cases with three alpha domains) and non-covalently associated β2 microglobulin. Generally, MHC class II molecules are composed of two transmembrane glycoproteins, α and β, both of which typically cross the membrane. MHC molecules may include an effective portion of MHC that contains an antigen binding site or sites for binding peptides and sequences necessary for recognition by an appropriate antigen receptor. In some embodiments, the MHC class I molecule delivers cytosolic derived peptides to the cell surface where the MHC-peptide complex is bound by a T cell (e.g., typically CD8 + T cells, but in some cases CD4 + T cells). In some embodiments, MHC class II molecules deliver peptides derived from the vesicle system to the cell surface, wherein the peptides are typically bound by CD4 + T cell recognition. Generally, MHC molecules are encoded by a set of linked loci, which are collectively referred to as H-2 in mice and Human Leukocyte Antigen (HLA) in humans. Thus, human MHC may also be referred to as Human Leukocyte Antigen (HLA) in general.
The term "MHC-peptide complex" or "peptide-MHC complex" or variants thereof refers to complexes or associations of peptide antigens with MHC molecules, e.g. typically formed by non-covalent interactions of the peptides in the binding groove or cleft of the MHC molecule. In some embodiments, the MHC-peptide complex is present or displayed on the surface of a cell. In some embodiments, the MHC-peptide complex can be specifically recognized by an antigen receptor (e.g., a TCR-like CAR, or an antigen-binding portion thereof).
In some embodiments, peptides (e.g., peptide antigens or epitopes) of the polypeptides may be associated with MHC molecules, such as for recognition by antigen receptors. Typically, the peptide is derived from or based on a fragment of a longer biomolecule (e.g., a polypeptide or protein). In some embodiments, the peptide generally has a length of about 8 to about 24 amino acids. In some embodiments, the peptide is from or about 9 to 22 amino acids in length for recognition in MHC class II complexes. In some embodiments, the peptide is from or from about 8 to 13 amino acids in length for recognition in an MHC class I complex. In some embodiments, upon recognition of a peptide in the context of an MHC molecule (e.g., MHC-peptide complex), an antigen receptor (e.g., a TCR or TCR-like CAR) generates or triggers an activation signal to a T cell, inducing a T cell response, such as T cell proliferation, cytokine production, cytotoxic T cell response, or other response.
In some embodiments, TCR-like antibodies or antigen-binding portions are known or can be produced by known methods (see, e.g., U.S. published application nos. US2002/0150914; US2003/0223994; US2004/0191260; US2006/0034850; US2007/00992530; US20090226474; US 2009043679; and international application publication nos. WO 03/068201).
In some embodiments, antibodies or antigen-binding portions thereof that specifically bind to MHC-peptide complexes may be produced by immunizing a host with an effective amount of an immunogen comprising the particular MHC-peptide complex. In some cases, the peptide of the MHC-peptide complex is an epitope of an antigen capable of binding to MHC, such as a tumor antigen, such as a general tumor antigen, a myeloma antigen, or other antigen as described below. In some embodiments, an effective amount of an immunogen is then administered to the host for eliciting an immune response, wherein the immunogen remains in its three-dimensional form for a time sufficient to elicit an immune response directed against the three-dimensional presentation of the peptide in the binding groove of the MHC molecule. Serum collected from the host is then assayed to determine whether the desired antibodies recognizing the three-dimensional presentation of peptides in the binding groove of the MHC molecule are produced. In some embodiments, the antibodies produced can be evaluated to confirm that the antibodies can distinguish MHC-peptide complexes from MHC molecules alone, peptides of interest alone, and complexes of MHC with unrelated peptides. The desired antibody can then be isolated.
In some embodiments, antibodies or antigen-binding portions thereof that specifically bind to MHC-peptide complexes can be generated by employing an antibody library display method (e.g., phage antibody library). In some embodiments, phage display libraries of mutant Fab, scFv, or other antibody forms can be generated, for example, wherein members of the library are mutated at one or more residues of one or more CDRs. See, for example, U.S. patent application publication nos. US 20020150914, US20140294841; cohen CJ. Et al (2003) J mol. Recog. 16:324-332.
In some embodiments, the antigen is CD20. In some embodiments, the scFv comprises VH and VL derived from antibodies or antibody fragments specific for CD20. In some embodiments, the antibody or antibody fragment that binds CD20 is an antibody that is rituximab or is derived from rituximab, such as rituximab scFv.
In some embodiments, the antigen is CD22. In some embodiments, the scFv comprises VH and VL derived from antibodies or antibody fragments specific for CD22. In some embodiments, the antibody or antibody fragment that binds CD22 is an antibody that is m971 or derived from m971, e.g., is an m971 scFv.
In some embodiments, the chimeric antigen receptor comprises an extracellular portion comprising an antibody or antibody fragment. In some aspects, the chimeric antigen receptor comprises an extracellular portion comprising an antibody or fragment and an intracellular signaling domain. In some embodiments, the antibody or fragment comprises an scFv.
In some embodiments, the antibody portion of the recombinant receptor (e.g., CAR) further comprises at least a portion of an immunoglobulin constant region, such as a hinge region (e.g., an IgG4 hinge region) and/or a CH1/CL and/or Fc region. In some embodiments, the constant region or portion is a constant region or portion of a human IgG (e.g., igG4 or IgG 1). In some aspects, the portion of the constant region serves as a spacer region between the antigen recognition component (e.g., scFv) and the transmembrane domain. The length of the spacer may provide enhanced cellular responsiveness after antigen binding compared to the absence of the spacer. Exemplary spacers include, but are not limited to, those described in the following documents: hudecek et al (2013) Clin.cancer Res.,19:3153; international patent application publication No. WO 2014031687, U.S. patent No. 8,822,647 or published application No. US 2014/0271635.
In some embodiments, the constant region or portion is a constant region or portion of a human IgG (e.g., igG4 or IgG 1). In some embodiments, the spacer has the sequence ESKYGPPCPPCP (as shown in SEQ ID NO: 69) and is encoded by the sequence shown in SEQ ID NO: 70. In some embodiments, the spacer has the sequence shown as SEQ ID NO. 71. In some embodiments, the spacer has the sequence shown as SEQ ID NO. 72. In some embodiments, the constant region or portion is a constant region or portion of IgD. In some embodiments, the spacer is part of an immunoglobulin constant region that is or comprises a hinge sequence. In some embodiments, the spacer has the sequence shown as SEQ ID NO. 73. In some embodiments, the spacer has an amino acid sequence that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to any one of SEQ ID NOs 69, 70, 71, 72 or 73. In some embodiments, the spacer has the sequence shown in SEQ ID NOS.74-82. In some embodiments, the spacer has an amino acid sequence that exhibits at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to any one of SEQ ID NOs 74-82.
In some embodiments, the antigen receptor comprises an intracellular domain linked directly or indirectly to an extracellular domain. In some embodiments, the chimeric antigen receptor comprises a transmembrane domain that connects an extracellular domain and an intracellular signaling domain. In some embodiments, the intracellular signaling domain comprises ITAM. For example, in some aspects, an antigen recognition domain (e.g., an extracellular domain) is typically linked to one or more intracellular signaling components (e.g., signaling components that mimic activation by an antigen receptor complex (e.g., a TCR complex) (in the case of a CAR) and/or signaling by another cell surface receptor. In some embodiments, the chimeric receptor comprises a transmembrane domain linked or fused between an extracellular domain (e.g., scFv) and an intracellular signaling domain. Thus, in some embodiments, an antigen binding component (e.g., an antibody) is linked to one or more transmembrane domains and intracellular signaling domains.
In one embodiment, a transmembrane domain is used that naturally associates with one of the domains in the receptor (e.g., CAR). In some cases, the transmembrane domains are selected or modified by amino acid substitutions to avoid binding of such domains to transmembrane domains of the same or different surface membrane proteins to minimize interactions with other members of the receptor complex.
In some embodiments, the transmembrane domain is derived from a natural or synthetic source. When the source is natural, in some aspects, the domain may be derived from any membrane-bound protein or transmembrane protein. The transmembrane regions include those derived from (i.e., comprising at least one or more of the transmembrane regions of): the α, β or ζ chain of the T cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154. Alternatively, in some embodiments, the transmembrane domain is synthetic. In some aspects, the synthetic transmembrane domain comprises predominantly hydrophobic residues, such as leucine and valine. In some aspects, triplets of phenylalanine, tryptophan and valine will be found at each end of the synthetic transmembrane domain. In some embodiments, the connection is through a linker, spacer and/or one or more transmembrane domains. In some aspects, the transmembrane domain comprises a transmembrane portion of CD 28.
In some embodiments, the extracellular domain and the transmembrane domain may be directly or indirectly linked. In some embodiments, the extracellular domain and the transmembrane domain are connected by a spacer (any spacer as described herein). In some embodiments, the receptor contains an extracellular portion of a molecule from which the transmembrane domain is derived, such as a CD28 extracellular portion.
Intracellular signaling domains include those that mimic or approximate: signaling through natural antigen receptors, signaling through a combination of such receptors with co-stimulatory receptors, and/or signaling through co-stimulatory receptors alone. In some embodiments, there is a short oligopeptide or polypeptide linker, e.g., a linker between 2 and 10 amino acids in length (e.g., a glycine and serine containing linker, e.g., a glycine-serine duplex), and a linkage is formed between the transmembrane domain and cytoplasmic signaling domain of the CAR.
In some aspects, T cell activation is described as being mediated by two classes of cytoplasmic signaling sequences: those that initiate antigen-dependent primary activation by TCRs (primary cytoplasmic signaling sequences), and those that act in an antigen-independent manner to provide secondary or costimulatory signals (secondary cytoplasmic signaling sequences). In some aspects, the CAR includes one or both of such signaling components.
Receptors (e.g., CARs) typically include at least one or more intracellular signaling components. In some aspects, the CAR comprises a primary cytoplasmic signaling sequence that modulates primary activation of the TCR complex. The primary cytoplasmic signaling sequence that acts in a stimulatory manner may contain a signaling motif (which is referred to as an immunoreceptor tyrosine activation motif or ITAM). Examples of primary cytoplasmic signaling sequences containing ITAM include those derived from the cd3ζ chain, fcrγ, cd3γ, cd3δ, and cd3ε. In some embodiments, the cytoplasmic signaling molecule in the CAR contains a cytoplasmic signaling domain derived from cd3ζ, a portion or sequence thereof.
In some embodiments, the receptor comprises an intracellular component of the TCR complex, such as a TCR CD3 chain, e.g., a cd3ζ chain, that mediates T cell activation and cytotoxicity. Thus, in some aspects, the antigen binding portion is linked to one or more cell signaling modules. In some embodiments, the cell signaling module comprises a CD3 transmembrane domain, a CD3 intracellular signaling domain, and/or other CD3 transmembrane domain. In some embodiments, the receptor (e.g., CAR) further comprises a portion of one or more additional molecules (e.g., fc receptor gamma, CD8, CD4, CD25, or CD 16). For example, in some aspects, the CAR or other chimeric receptor comprises a chimeric molecule between CD3-zeta (CD 3- ζ) or Fc receptor γ and CD8, CD4, CD25, or CD 16.
In some embodiments, upon attachment of a CAR or other chimeric receptor, the cytoplasmic domain or intracellular signaling domain of the receptor activates at least one of the normal effector functions or responses of an immune cell (e.g., a T cell engineered to express the CAR). For example, in some contexts, the CAR induces a function of a T cell, such as cytolytic activity or T helper cell activity, such as secretion of cytokines or other factors. In some embodiments, a truncated portion of the intracellular signaling domain of the antigen receptor component or co-stimulatory molecule (e.g., if it transduces an effector function signal) is used in place of the intact immunostimulatory chain. In some embodiments, one or more intracellular signaling domains include cytoplasmic sequences of T Cell Receptors (TCRs), and in some aspects also include those cytoplasmic sequences of co-receptors that cooperate with such receptors in the natural environment to initiate signal transduction upon antigen receptor engagement.
In the case of native TCRs, complete activation typically requires not only signaling via the TCR, but also a co-stimulatory signal. Thus, in some embodiments, to facilitate complete activation, components for generating secondary or co-stimulatory signals are also included in the CAR. In other embodiments, the CAR does not include a component for generating a co-stimulatory signal. In some aspects, the additional CAR is expressed in the same cell and provides a component for generating a secondary or co-stimulatory signal.
In some embodiments, the chimeric antigen receptor comprises an intracellular domain of a T cell costimulatory molecule. In some embodiments, the CAR comprises a signaling domain and/or transmembrane portion of a co-stimulatory receptor (e.g., CD28, 4-1BB, OX40, DAP10, and ICOS). In some aspects, the same CAR includes both an activating component and a co-stimulatory component. In some embodiments, the chimeric antigen receptor comprises an intracellular domain derived from a T cell costimulatory molecule, or a functional variant thereof, such as located between the transmembrane domain and the intracellular signaling domain. In some aspects, the T cell costimulatory molecule is CD28 or 41BB.
In some embodiments, the activation domain is included within one CAR, and the co-stimulatory component is provided by another CAR that recognizes another antigen. In some embodiments, the CAR comprises an activated or stimulated CAR, a co-stimulated CAR (see WO 2014/055668), all expressed on the same cell. In some aspects, the cell includes one or more stimulating or activating CARs and/or co-stimulating CARs. In some embodiments, the cells further comprise an inhibitory CAR (iCAR, see Fedorov et al, sci.tranl.medicine, 5 (215) (month 12 2013), such as a CAR that recognizes an antigen other than an antigen associated with and/or characteristic of a disease or disorder, wherein an activation signal delivered by a disease-targeted CAR is reduced or inhibited by binding of the inhibitory CAR to its ligand, e.g., to reduce off-target effects.
In some embodiments, the two receptors induce activation and inhibitory signals to the cell, respectively, such that the attachment of one receptor to its antigen activates the cell or induces a response, but the attachment of the second inhibitory receptor to its antigen induces a signal that inhibits or attenuates the response. An example is the combination of an activating CAR with an Inhibitory CAR (iCAR). For example, such a strategy may be used, for example, to reduce the likelihood of off-target effects in the context in which activating a CAR binds to an antigen that is expressed in a disease or disorder but is also expressed on normal cells, and an inhibitory receptor binds to a separate antigen that is expressed on normal cells but is not expressed on cells of the disease or disorder.
In some aspects, the chimeric receptor is or includes an inhibitory CAR (e.g., iCAR) and includes an intracellular component that attenuates or inhibits an immune response (e.g., ITAM in a cell and/or co-stimulates a promoted response). Examples of such intracellular signaling components are those found on immune checkpoint molecules, including PD-1, CTLA4, LAG3, BTLA, OX2R, TIM-3, TIGIT, LAIR-1, PGE2 receptors, EP2/4 adenosine receptors (including A2 AR). In some aspects, the engineered cells include an inhibitory CAR comprising or derived from a signaling domain of such an inhibitory molecule, such that it is used to attenuate cellular responses induced, for example, by activating and/or co-stimulating the CAR.
In certain embodiments, the intracellular signaling domain comprises a CD28 transmembrane and signaling domain linked to a CD3 (e.g., CD3- ζ) intracellular domain. In some embodiments, the intracellular signaling domain comprises a chimeric CD28 and CD137 (4-1 BB, TNFRSF9) co-stimulatory domain linked to a CD3 zeta intracellular domain.
In some embodiments, the CAR encompasses one or more (e.g., two or more) co-stimulatory domains and an activation domain (e.g., a primary activation domain) in the cytoplasmic portion. Exemplary CARs include intracellular components of CD 3-zeta, CD28, and 4-1 BB.
In some embodiments, the antigen receptor further comprises a label, and/or the cell expressing the CAR or other antigen receptor further comprises a surrogate marker, such as a cell surface marker, which can be used to confirm that the cell is transduced or engineered to express the receptor. In some aspects, the marker comprises all or part (e.g., a truncated form) of CD34, NGFR, or an epidermal growth factor receptor, such as a truncated form of such a cell surface receptor (e.g., tgfr). In some embodiments, the nucleic acid encoding the marker is operably linked to a polynucleotide encoding a linker sequence (e.g., a cleavable linker sequence, such as T2A). For example, the tag and optionally the linker sequence may be any as disclosed in published patent application number WO 2014031687. For example, the marker may be truncated EGFR (tEGFR), which is optionally linked to a linker sequence, such as a T2A cleavable linker sequence.
Exemplary polypeptides of truncated EGFR (e.g., tEGFR) comprise the amino acid sequence shown in SEQ ID NO:2 or 3 or an amino acid sequence exhibiting at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO:2 or 3. Exemplary T2A linker sequences comprise the amino acid sequence shown in SEQ ID NO. 1 or 4 or an amino acid sequence exhibiting at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO. 1 or 4.
In some embodiments, the marker is a molecule (e.g., a cell surface protein) or portion thereof that is not found naturally on a T cell or is not found naturally on a T cell surface. In some embodiments, the molecule is a non-self molecule, e.g., a non-self protein, i.e., a molecule that is not recognized as "self" by the host immune system of the adoptively transferred cell.
In some embodiments, the marker does not play any therapeutic role and/or does not produce a role other than that used as a genetically engineered marker (e.g., for selecting successfully engineered cells). In other embodiments, the marker may be a therapeutic molecule or a molecule that otherwise exerts a desired effect, such as a ligand of a cell encountered in vivo, such as a co-stimulatory or immune checkpoint molecule, to enhance and/or attenuate the response of the cell upon adoptive transfer and upon encountering the ligand.
In some cases, the CAR is referred to as a first, second, and/or third generation CAR. In some aspects, the first generation CAR is a CAR that provides only CD3 chain-induced signaling upon antigen binding; in some aspects, the second generation CAR is a CAR that provides such a signal and a co-stimulatory signal, such as a signal comprising an intracellular signaling domain from a co-stimulatory receptor such as CD28 or CD 137; in some aspects, the third generation CAR is a CAR comprising multiple co-stimulatory domains of different co-stimulatory receptors.
For example, in some embodiments, the CAR contains an antibody (e.g., an antibody fragment, such as scFv) that is specific for an antigen (including any of those described), a transmembrane domain that is or contains a transmembrane portion of CD28 or a functional variant thereof, and an intracellular signaling domain that contains a signaling portion of CD28 or a functional variant thereof and a signaling portion of cd3ζ or a functional variant thereof. In some embodiments, the CAR contains an antibody (e.g., an antibody fragment, such as scFv) that is specific for an antigen (including any of those described), a transmembrane domain that is or contains a transmembrane portion of CD28 or a functional variant thereof, and an intracellular signaling domain that contains a signaling portion of 4-1BB or a functional variant thereof and a signaling portion of cd3ζ or a functional variant thereof. In some such embodiments, the receptor further comprises a spacer, such as a hinge-only spacer, comprising a portion (e.g., an Ig hinge, e.g., an IgG4 hinge) of an Ig molecule (e.g., a human Ig molecule).
In some embodiments, the transmembrane domain of a recombinant receptor (e.g., CAR) is or comprises the transmembrane domain of human CD28 (e.g., accession number P01747.1) or a variant thereof, such as a transmembrane domain comprising the amino acid sequence set forth in SEQ ID No. 83 or an amino acid sequence exhibiting at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID No. 83; in some embodiments, the transmembrane domain-containing portion of the recombinant receptor comprises the amino acid sequence set forth in SEQ ID NO 84 or an amino acid sequence having at least or about 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity thereto.
In some embodiments, one or more intracellular signaling components of a recombinant receptor (e.g., CAR) contain an intracellular co-stimulatory signaling domain of human CD28 or a functional variant or portion thereof, such as a domain having LL to GG substitutions at positions 186-187 of the native CD28 protein. For example, the intracellular signaling domain may comprise the amino acid sequence shown in SEQ ID No. 85 or 86 or an amino acid sequence exhibiting at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID No. 85 or 86. In some embodiments, the intracellular domain comprises the intracellular co-stimulatory signaling domain of 4-1BB (e.g., accession number Q07011.1), or a functional variant or portion thereof, such as the amino acid sequence shown in SEQ ID NO:87 or an amino acid sequence exhibiting at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO: 87.
In some embodiments, the intracellular signaling domain of a recombinant receptor (e.g., CAR) comprises a human CD3 zeta stimulating signaling domain or a functional variant thereof, such as the cytoplasmic domain of 112 AA of subtype 3 of human CD3 zeta (accession number: P20963.2) or as in U.S. patent No.: a CD3 zeta signaling domain described in 7,446,190 or U.S. patent No. 8,911,993. For example, in some embodiments, the intracellular signaling domain comprises the amino acid sequence shown in SEQ ID No. 88, 89, or 90 or an amino acid sequence exhibiting at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID No. 88, 89, or 90.
In some aspects, the spacer contains only hinge regions of IgG, such as only hinge of IgG4 or IgG1, and only hinge spacer as shown in SEQ ID NO: 69. In other embodiments, the spacer is or comprises an Ig hinge, such as an IgG 4-derived hinge, optionally linked to a CH2 and/or CH3 domain. In some embodiments, the spacer is an Ig hinge, e.g., an IgG4 hinge, linked to CH2 and CH3 domains, as shown in SEQ ID NO: 72. In some embodiments, the spacer is an Ig hinge, e.g., an IgG4 hinge, linked only to the CH3 domain, as shown in SEQ ID NO: 71. In some embodiments, the spacer is or comprises a glycine-serine rich sequence or other flexible linker, such as known flexible linkers.
For example, in some embodiments, the CAR comprises an antibody (e.g., an antibody fragment, including an scFv), a spacer (e.g., a spacer comprising a portion of an immunoglobulin molecule (e.g., a hinge region and/or one or more constant regions of a heavy chain molecule), such as a spacer comprising an Ig hinge, a transmembrane domain comprising all or a portion of a CD 28-derived transmembrane domain, a CD 28-derived intracellular signaling domain, and a CD3 zeta signaling domain. In some embodiments, the CAR comprises an antibody or fragment (e.g., scFv), a spacer (e.g., any spacer comprising an Ig hinge), a CD 28-derived transmembrane domain, a 4-1 BB-derived intracellular signaling domain, and a cd3ζ -derived signaling domain.
Exemplary surrogate markers may include truncated forms of cell surface polypeptides, such as nonfunctionalAnd does not transduce or is incapable of transducing signals or signals that are normally transduced by a full length form of a cell surface polypeptide, and/or truncated forms that are not internalized or are incapable of internalization. Exemplary truncated cell surface polypeptides include truncated forms of growth factors or other receptors, such as truncated human epidermal growth factor receptor 2 (tper 2), truncated epidermal growth factor receptor (tgfr, exemplary tgfr sequences shown in 2 or 3), or Prostate Specific Membrane Antigen (PSMA), or modified forms thereof. tEGFR may contain a polypeptide consisting of the antibody cetuximab Or other therapeutic anti-EGFR antibodies or binding molecules, which can be used to identify or select cells that have been engineered to express the tgfr construct and the encoded exogenous protein, and/or to eliminate or isolate cells that express the encoded exogenous protein. See U.S. patent No. 8,802,374 and Liu et al, nature biotech.2016, month 4; 34 (4):430-434). In some aspects, the marker (e.g., surrogate marker) comprises all or part (e.g., truncated form) of CD34, NGFR, CD19, or truncated CD19 (e.g., truncated non-human CD 19) or an epidermal growth factor receptor (e.g., tgfr). In some embodiments, the label is or comprises a fluorescent protein, such as Green Fluorescent Protein (GFP), enhanced Green Fluorescent Protein (EGFP) (e.g., superfolder GFP (sfGFP)), red Fluorescent Protein (RFP) (e.g., tdTomato, mCherry, mStrawberry, asRed, dsRed or DsRed 2), cyan Fluorescent Protein (CFP), blue-green fluorescent protein (BFP), enhanced Blue Fluorescent Protein (EBFP), and Yellow Fluorescent Protein (YFP), and variants thereof, including species variants, monomer variants, and codon optimized and/or enhanced variants of fluorescent protein. In some embodiments, the label is or comprises an enzyme (e.g., luciferase), the lacZ gene from E.coli, alkaline phosphatase, secreted Embryonic Alkaline Phosphatase (SEAP), chloramphenicol Acetyl Transferase (CAT). Exemplary luminescent reporter genes include luciferase (luc), beta-galactosidase, chloramphenicol Acetyl Transferase (CAT), beta-Glucuronidase (GUS), or variants thereof.
In some embodiments, the marker is a resistance marker or a selection marker. In some embodiments, the resistance marker or selectable marker is or comprises a polypeptide that confers resistance to an exogenous agent or drug. In some embodiments, the resistance marker or selectable marker is an antibiotic resistance gene. In some embodiments, the resistance marker or selectable marker is an antibiotic resistance gene that confers antibiotic resistance to mammalian cells. In some embodiments, the resistance marker or selectable marker is or comprises a puromycin resistance gene, a hygromycin resistance gene, a blasticidin resistance gene, a neomycin resistance gene, a geneticin resistance gene, or a bleomycin resistance gene, or a modified version thereof.
In some embodiments, the nucleic acid encoding the marker is operably linked to a polynucleotide encoding a linker sequence (e.g., a cleavable linker sequence, e.g., T2A). For example, the tag and optionally the linker sequence may be as in PCT publication No. WO 2014031687.
In some embodiments, the nucleic acid molecule encoding such CAR construct further comprises, e.g., downstream of the sequence encoding the CAR, a sequence encoding a T2A ribosome-hopping element and/or a tgfr sequence. In some embodiments, the sequence encodes a T2A ribosome jump element as set forth in SEQ ID NO. 1 or 4, or an amino acid sequence exhibiting at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO. 1 or 4.
In some embodiments, T cells expressing an antigen receptor (e.g., CAR) can also be generated to express truncated EGFR (tgfr) as a non-immunogenic selection epitope (e.g., by introducing constructs encoding CAR and tgfr separated by a T2A ribosomal switch to express both proteins from the same construct), which can then be used as a marker to detect such cells (see, e.g., us patent No. 8,802,374). In some embodiments, the sequence encodes a tEGFR sequence shown in SEQ ID NO. 2 or 3, or an amino acid sequence exhibiting at least 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more sequence identity to SEQ ID NO. 2 or 3. In some cases, peptides such as T2A may cause ribosomes to skip synthesis of peptide bonds at the C-terminus of the 2A element (ribosome skipping), resulting in separation between the 2A sequence end and the next peptide downstream (see, e.g., de Felipe. Genetic Vaccines and Ther.2:13 (2004) and de Felipe et al Traffic5:616-626 (2004)). Many 2A elements are known. Examples of 2A sequences that may be used in the methods and nucleic acids disclosed herein include, but are not limited to, 2A sequences from: foot-and-mouth disease virus (F2A, e.g., SEQ ID NO: 8), equine rhinitis A virus (E2A, e.g., SEQ ID NO: 7), leptopetalum album beta tetrad virus (T2A, e.g., SEQ ID NO:1 or 4), and porcine teschovirus-1 (P2A, e.g., SEQ ID NO:5 or 6), as described in U.S. patent publication No. 20070116690.
Recombinant receptors (e.g., CARs) expressed by cells administered to a subject typically recognize or specifically bind to molecules that are expressed in, associated with, and/or specific for the disease or disorder being treated or cells thereof. Upon specific binding to a molecule (e.g., an antigen), the receptor typically delivers an immunostimulatory signal (e.g., an ITAM-transduced signal) into the cell, thereby facilitating an immune response that targets the disease or disorder. For example, in some embodiments, the cell expresses a CAR that specifically binds to an antigen expressed by or associated with a cell or tissue of the disease or disorder.
B. Chimeric autoantibody receptors (CAAR)
In some embodiments, the recombinant receptor is a chimeric autoantibody receptor (CAAR). In some embodiments, the CAAR binds (e.g., specifically binds) or recognizes an autoantibody. In some embodiments, cells expressing CAAR (e.g., T cells engineered to express CAAR) may be used to bind to and kill cells expressing autoantibodies, rather than cells expressing normal antibodies. In some embodiments, the CAAR expressing cells may be used to treat an autoimmune disease, such as an autoimmune disease, associated with the expression of an autoantigen. In some embodiments, CAAR expressing cells may target B cells that ultimately produce and display autoantibodies on their cell surfaces, marking these B cells as disease-specific targets for therapeutic intervention. In some embodiments, CAAR expressing cells may be used to target disease-causing B cells by using antigen specific chimeric autoantibody receptors to effectively target and kill pathogenic B cells in autoimmune diseases. In some embodiments, the recombinant receptor is CAAR, as described in any of the U.S. patent application publication US 2017/0051035.
In some embodiments, the CAAR comprises an autoantibody binding domain, a transmembrane domain, and one or more intracellular signaling domains or domains (also interchangeably referred to as cytoplasmic signaling domains or regions). In some embodiments, the intracellular signaling region comprises an intracellular signaling domain. In some embodiments, the intracellular signaling domain is or comprises a primary signaling domain, a signaling domain capable of stimulating and/or inducing a primary activation signal in a T cell, a signaling domain of a T Cell Receptor (TCR) component (e.g., an intracellular signaling domain or region of a CD3-Zeta (CD 3 Zeta) chain or a functional variant or signaling portion thereof), and/or a signaling domain comprising an immune receptor tyrosine activation motif (ITAM).
In some embodiments, the autoantibody binding domain comprises an autoantigen or fragment thereof. The choice of autoantigen may depend on the type of autoantibody targeted. For example, an autoantigen may be selected as a result of its recognition of an autoantibody on a target cell (e.g., B cell) associated with a particular disease state (e.g., an autoimmune disease, such as an autoantibody-mediated autoimmune disease). In some embodiments, the autoimmune disease comprises Pemphigus Vulgaris (PV). Exemplary autoantigens include desmosomal mucin 1 (Dsg 1) and Dsg3.
C.T cell receptor (TCR)
In some embodiments, an engineered cell (e.g., a T cell) is provided that expresses a T Cell Receptor (TCR) or antigen binding portion thereof that recognizes a peptide epitope or T cell epitope of a target polypeptide (e.g., an antigen of a tumor, virus, or autoimmune protein).
In some embodiments, a "T cell receptor" or "TCR" is a molecule or antigen-binding portion thereof that contains variable alpha and beta chains (also referred to as TCR alpha and TCR beta, respectively) or variable gamma and delta chains (also referred to as TCR alpha and TCR beta, respectively), and which is capable of specifically binding to peptides that bind to MHC molecules. In some embodiments, the TCR is in the αβ form. Generally, TCRs in the form of αβ and γδ are generally similar in structure, but T cells expressing them may have different anatomical locations or functions. TCRs can be found on the surface of cells or in soluble form. Typically, TCRs are found on the surface of T cells (or T lymphocytes), where they are generally responsible for recognizing antigens bound to Major Histocompatibility Complex (MHC) molecules.
The term "TCR" is understood to encompass an intact TCR, as well as antigen-binding portions or antigen-binding fragments thereof, unless otherwise indicated. In some embodiments, the TCR is a complete or full length TCR, including TCRs in αβ form or γδ form. In some embodiments, the TCR is an antigen-binding portion that is less than a full-length TCR but binds to a specific peptide that binds in an MHC molecule (e.g., to an MHC-peptide complex). In some cases, the antigen binding portion or fragment of the TCR may contain only a portion of the structural domain of the full length or complete TCR, but still be able to bind to a peptide epitope (e.g., MHC-peptide complex) that binds to the complete TCR. In some cases, the antigen binding portion contains the variable domains of the TCR (e.g., the variable alpha and beta chains of the TCR) sufficient to form a binding site for binding to a particular MHC-peptide complex. Typically, the variable chain of a TCR contains complementarity determining regions involved in the recognition of peptides, MHC and/or MHC-peptide complexes.
In some embodiments, the variable domain of the TCR contains hypervariable loops or Complementarity Determining Regions (CDRs), which are generally the major contributors to antigen recognition and binding capacity and specificity. In some embodiments, the CDRs of a TCR, or a combination thereof, form all or substantially all of the antigen binding sites of a given TCR molecule. The individual CDRs within the variable region of the TCR chain are typically separated by Framework Regions (FRs) which generally exhibit lower variability between TCR molecules than CDRs (see, e.g., jores et al, proc. Nat' l Acad. Sci. U.S. A.87:9138,1990; chothia et al, EMBO J.7:3745,1988; see also Lefranc et al, dev. Comp. Immunol.27:55,2003). In some embodiments, CDR3 is the primary CDR responsible for antigen binding or specificity, or the most important of the three CDRs for antigen recognition and/or for interaction with the processed peptide portion of the peptide-MHC complex at a given TCR variable region. In some cases, CDR1 of the alpha chain may interact with the N-terminal portion of certain antigenic peptides. In some cases, CDR1 of the β chain may interact with the C-terminal portion of the peptide. In some cases, CDR2 has the strongest effect on interaction or recognition with the MHC portion of the MHC-peptide complex or is primarily responsible for the CDR. In some embodiments, the variable region of the β chain may contain other hypervariable regions (CDR 4 or HVR 4) that are normally involved in superantigen binding rather than antigen recognition (Kotb (1995) Clinical Microbiology Reviews, 8:411-426).
In some embodiments, the TCR can also contain constant domains, transmembrane domains, and/or short cytoplasmic tails (see, e.g., janeway et al, immunobiology: the Immune Systemin Health and Disease, 3 rd edition, current Biology Publications, page 4: 33,1997). In some aspects, each chain of the TCR can have an N-terminal immunoglobulin variable domain, an immunoglobulin constant domain, a transmembrane region, and a short cytoplasmic tail at the C-terminus. In some embodiments, the TCR is associated with a invariant protein of the CD3 complex involved in mediating signal transduction.
In some embodiments, the TCR chain comprises one or more constant domains. For example, the extracellular portion of a given TCR chain (e.g., an alpha chain or a beta chain) may contain two immunoglobulin-like domains adjacent to the cell membrane, such as a variable domain (e.g., vα or vβ; typically amino acids 1 to 116 based on Kabat numbering, kabat et al, "Sequences of Proteins of Immunological Interest", usdept. Health and Human Services, public Health Service National Institutes of Health,1991, version 5) and a constant domain (e.g., an alpha chain constant domain or cα, typically positions 117 to 259 of a Kabat numbering-based chain; or a beta chain constant domain or cβ, typically positions 117 to 295 of a Kabat numbering-based chain). For example, in some cases, the extracellular portion of a TCR formed by two chains contains two membrane proximal constant domains and two membrane distal variable domains, wherein the variable domains each contain CDRs. The constant domain of the TCR may contain a short linking sequence in which the cysteine residues form a disulfide bond, thereby linking the two chains of the TCR. In some embodiments, the TCR may have additional cysteine residues in each of the α and β chains, such that the TCR contains two disulfide bonds in the constant domain.
In some embodiments, the TCR chain comprises a transmembrane domain. In some embodiments, the transmembrane domain is positively charged. In some cases, the TCR chain contains a cytoplasmic tail. In some cases, the structure allows the TCR to associate with other molecules (e.g., CD3 and its subunits). For example, TCRs containing constant domains and transmembrane regions can anchor proteins in the cell membrane and associate with a constant subunit of a CD3 signaling device or complex. The intracellular tail of the CD3 signaling subunits (e.g., the cd3γ, cd3δ, cd3ε, and cd3ζ chains) contain one or more immune receptor tyrosine activation motifs or ITAMs involved in the signaling capacity of the TCR complex.
In some embodiments, the TCR may be a heterodimer of two chains α and β (or optionally γ and δ), or it may be a single chain TCR construct. In some embodiments, the TCR is a heterodimer comprising two separate chains (alpha and beta chains or gamma and delta chains) linked by, for example, one or more disulfide bonds.
In some embodiments, TCRs may be generated from one or more known TCR sequences (e.g., sequences of vα, β chains) whose substantially full-length coding sequences are readily available. Methods for obtaining full length TCR sequences (including V chain sequences) from cellular sources are well known. In some embodiments, the nucleic acid encoding the TCR may be obtained from a variety of sources, such as by Polymerase Chain Reaction (PCR) amplification of the nucleic acid encoding the TCR within or isolated from one or more given cells, or by synthesis of publicly available TCR DNA sequences.
In some embodiments, the TCR is obtained from a biological source, such as from a cell (e.g., from a T cell (e.g., a cytotoxic T cell)), a T cell hybridoma, or other publicly available source. In some embodiments, T cells may be obtained from cells isolated in vivo. In some embodiments, the TCR is a thymus-selected TCR. In some embodiments, the TCR is a neoepitope-restricted TCR. In some embodiments, the T cell may be a cultured T cell hybridoma or clone. In some embodiments, the TCR, or an antigen-binding portion thereof, or an antigen-binding fragment thereof, may be synthetically generated based on knowledge of TCR sequences.
In some embodiments, the TCR is generated from a TCR identified or selected by screening a candidate TCR library for a target polypeptide antigen or target T cell epitope thereof. TCR libraries can be generated by expanding V alpha and V beta libraries from T cells isolated from a subject, including cells present in PBMCs, spleen, or other lymphoid organs. In some cases, T cells may be expanded from Tumor Infiltrating Lymphocytes (TILs). In some embodiments, TCR libraries may be generated from cd4+ or cd8+ T cells. In some embodiments, the TCR may be amplified from a T cell source (i.e., a normal TCR library) of a normal or healthy subject. In some embodiments, TCRs may be amplified from a T cell source in a subject with disease, i.e., a library of diseased TCRs. In some embodiments, libraries of V.alpha.and V.beta.genes are amplified using degenerate primers, such as by performing RT-PCR in samples (e.g., T cells) obtained from humans. In some embodiments, scTv libraries may be assembled from natural vα and vβ libraries, wherein amplified products are cloned or assembled to be separated by linkers. Depending on the subject and the source of the cells, the library may be HLA allele specific. Alternatively, in some embodiments, a TCR library can be generated by mutagenesis or diversification of a parent or scaffold TCR molecule. In some aspects, the TCR is subjected to directed evolution, e.g., of the alpha or beta chain, such as by mutagenesis. In some aspects, specific residues within the CDRs of the TCR are altered. In some embodiments, the selected TCR can be modified by affinity maturation. In some embodiments, antigen-specific T cells may be selected, such as by screening, to assess CTL activity against the peptide. In some aspects, TCRs present on antigen-specific T cells, for example, can be selected, such as by binding activity (e.g., a particular affinity or avidity) to an antigen.
In some embodiments, the TCR, or antigen-binding portion thereof, is modified or engineered. In some embodiments, directed evolution methods are used to generate TCRs with altered properties, such as having higher affinity for a particular MHC-peptide complex. In some embodiments, directed evolution is achieved by display methods including, but not limited to, yeast display (Holler et al (2003) Nat Immunol,4,55-62; holler et al (2000) Proc Natl Acad Sci U S A,97,5387-92); phage display (Li et al (2005) Nat Biotechnol,23,349-54) or T cell display (Chervin et al (2008) J Immunol Methods,339,175-84). In some embodiments, the display pathway involves engineering or modifying a known parent or reference TCR. For example, in some cases, a wild-type TCR may be used as a template for generating a mutagenized TCR in which one or more residues of the CDRs are mutated and mutants are selected that have the desired altered properties (e.g., higher affinity for the desired target antigen).
In some embodiments, the peptide used to produce or generate the target polypeptide of the TCR of interest is known or can be readily identified. In some embodiments, peptides suitable for use in generating a TCR or antigen-binding portion can be determined based on the presence of HLA restriction motifs in a target polypeptide of interest (target polypeptide as described below). In some embodiments, the peptides are identified using available in silico predictive models. In some embodiments, such models include, but are not limited to, proPred1 (Singh and Raghava (2001) Bioinformation 17 (12): 1236-1237) and SYFPEITHI (see Schulter et al (2007) Immunoinformatics Methods in Molecular Biology,409 (1): 75-93 2007) for predicting MHC class I binding sites. In some embodiments, the MHC restriction epitope is HLA-A0201, which is expressed in about 39% to 46% of all caucasians, and thus represents a suitable choice of MHC antigen for the preparation of a TCR or other MHC-peptide binding molecule.
The HLA-A0201 binding motifs and cleavage sites of proteasomes and immunoproteasome using computer predictive models are known. Such models for predicting MHC class I binding sites include, but are not limited to, proPred1 (described in more detail in Singh and Raghava, proPred: prediction of HLA-DR binding sites. BIOINFORMATICS17 (12): 1236-1237 2001) and SYFPEITHI (see Schulter et al SYFPEITHIDatabase for Searching and T-Cell Epitope prediction, immunoinformatics Methods in Molecular Biology, volume 409 (1): 75-93 2007).
In some embodiments, the TCR, or antigen-binding portion thereof, may be a recombinantly produced native protein or a mutant form thereof (in which one or more characteristics (e.g., binding characteristics) have been altered). In some embodiments, the TCR may be derived from one of a variety of animal species, such as human, mouse, rat, or other mammal. TCRs may be cell-bound or in soluble form. In some embodiments, for the purposes of the provided methods, the TCR is in a cell-bound form expressed on the surface of a cell.
In some embodiments, the TCR is a full length TCR. In some embodiments, the TCR is an antigen-binding moiety. In some embodiments, the TCR is a dimeric TCR (dTCR). In some embodiments, the TCR is a single chain TCR (sc-TCR). In some embodiments, the dTCR or scTCR has a structure as described in WO 03/020763, WO 04/033685, WO 2011/044186.
In some embodiments, the TCR comprises a sequence corresponding to a transmembrane sequence. In some embodiments, the TCR does contain a sequence corresponding to a cytoplasmic sequence. In some embodiments, the TCR is capable of forming a TCR complex with CD 3. In some embodiments, any TCR (including dTCR or scTCR) may be linked to a signaling domain that produces an active TCR on the surface of a T cell. In some embodiments, the TCR is expressed on the surface of a cell.
In some embodiments, the dTCR comprises a first polypeptide (wherein the sequence corresponding to the TCR a chain variable region sequence is fused to the N-terminus of the sequence corresponding to the TCR a chain constant region extracellular sequence) and a second polypeptide (wherein the sequence corresponding to the TCR β chain variable region sequence is fused to the N-terminus of the sequence corresponding to the TCR β chain constant region extracellular sequence), the first and second polypeptides being linked by a disulfide bond. In some embodiments, the bond may correspond to a native interchain disulfide bond present in a native dimeric αβ TCR. In some embodiments, the interchain disulfide bonds are not present in a native TCR. For example, in some embodiments, one or more cysteines may be incorporated into the constant region extracellular sequence of a dTCR polypeptide pair. In some cases, both natural and non-natural disulfide bonds may be desired. In some embodiments, the TCR contains a transmembrane sequence to anchor to the membrane.
In some embodiments, the dTCR comprises a TCR a chain (comprising a variable a domain, a constant a domain, and a first dimerization motif attached to the C-terminus of the constant a domain) and a TCR β chain (comprising a variable β domain, a constant β domain, and a first dimerization motif attached to the C-terminus of the constant β domain), wherein the first and second dimerization motifs readily interact to form a covalent bond between an amino acid of the first dimerization motif and an amino acid of the second dimerization motif, thereby linking the TCR a chain to the TCR β chain.
In some embodiments, the TCR is a scTCR. In general, scTCR can be generated using known methods, see, e.g., soo Hoo, W.F. et al PNAS (USA) 89,4759 (1992); tulfing, C.and Pluckthun, A., J.mol.biol.242,655 (1994); kurucz, i. et al PNAS (USA) 90 3830 (1993); international publication PCT nos. WO 96/13593, WO 96/18105, WO 99/60120, WO 99/18129, WO 03/020763, WO 2011/044186; and Schluetter, C.J. et al J.mol.biol.256,859 (1996). In some embodiments, the scTCR contains an incorporated unnatural inter-chain disulfide bond to facilitate association of TCR chains (see, e.g., international publication PCT No. WO 03/020763). In some embodiments, the scTCR is a non-disulfide linked truncated TCR in which the heterologous leucine zipper fused to its C-terminus facilitates chain association (see, e.g., international publication PCT No. WO 99/60120). In some embodiments, the scTCR comprises a TCR a variable domain covalently linked to a TCR β variable domain through a peptide linker (see, e.g., international publication PCT No. WO 99/18129).
In some embodiments, the scTCR comprises a first segment comprising an amino acid sequence corresponding to a TCR α chain variable region, a second segment comprising an amino acid sequence corresponding to a TCR β chain variable region sequence (fused to the N-terminus of the amino acid sequence corresponding to a TCR β chain constant domain extracellular sequence), and a linker sequence connecting the C-terminus of the first segment to the N-terminus of the second segment.
In some embodiments, the scTCR comprises a first segment (which consists of an alpha chain variable region sequence fused to the N-terminus of an alpha chain extracellular constant domain sequence) and a second segment (which consists of a beta chain variable region sequence fused to the N-terminus of a sequence beta chain extracellular constant and transmembrane sequence), and optionally a linker sequence (which connects the C-terminus of the first segment to the N-terminus of the second segment).
In some embodiments, the scTCR comprises a first segment (which consists of a TCR β chain variable region sequence fused to the N-terminus of a β chain extracellular constant domain sequence) and a second segment (which consists of an a chain variable region sequence fused to the N-terminus of a sequence a chain extracellular constant and transmembrane sequence), and optionally a linker sequence (which connects the C-terminus of the first segment to the N-terminus of the second segment).
In some embodiments, the linker of the scTCR that connects the first and second TCR segments may be any linker capable of forming a single polypeptide chain while preserving TCR binding specificity. In some embodiments, the linker sequence may, for example, have the formula-P-AA-P-, wherein P is proline and AA represents an amino acid sequence, wherein the amino acids are glycine and serine. In some embodiments, the first and second segments are paired such that their variable region sequences are oriented for such binding. Thus, in some cases, the linker is of sufficient length to span the distance between the C-terminus of the first segment and the N-terminus of the second segment, or vice versa, but not so long as to block or reduce binding of the scTCR to the target ligand. In some embodiments, the linker may contain from 10 to 45 amino acids or from about 10 to about 45 amino acids, such as 10 to 30 amino acids or 26 to 41 amino acid residues, e.g., 29, 30, 31, or 32 amino acids. In some embodiments, the linker has the formula-PGGG- (SGGGG) 5-P-, wherein P is proline, G is glycine and S is serine (SEQ ID NO: 91). In some embodiments, the linker has the sequence GSADDAKKDAAKKDGKS (SEQ ID NO: 92)
In some embodiments, the scTCR contains a covalent disulfide bond that links a residue of an immunoglobulin region of a constant domain of an alpha chain to a residue of an immunoglobulin region of a constant domain of a beta chain. In some embodiments, there are no interchain disulfide bonds in the native TCR. For example, in some embodiments, one or more cysteines may be incorporated into the constant region extracellular sequences of the first and second segments of the scTCR polypeptide. In some cases, both natural and non-natural disulfide bonds may be desired.
In some embodiments of dTCR or scTCR containing an introduced interchain disulfide bond, no native disulfide bond is present. In some embodiments, another residue is substituted with one or more native cysteines that form a native interchain disulfide bond, such as a serine or alanine substitution. In some embodiments, the introduced disulfide bond may be formed by mutating non-cysteine residues on the first and second segments to cysteines. Exemplary unnatural disulfide bonds for TCRs are described in published international PCT publication No. WO 2006/000830.
In some embodiments, the TCR, or antigen-binding fragment thereof, exhibits an affinity for the target antigen that balances the binding constant at or between about 10 "5 and 10" 12M, and all individual values and ranges therein. In some embodiments, the target antigen is an MHC-peptide complex or ligand.
In some embodiments, one or more nucleic acids encoding a TCR (e.g., alpha and beta strands) can be amplified by PCR, cloning, or other suitable means, and cloned into one or more suitable expression vectors. The expression vector may be any suitable recombinant expression vector and may be used to transform or transfect any suitable host. Suitable vectors include those designed for propagation and amplification or for expression or both, such as plasmids and viruses.
In some embodiments, the vector may be a series of vectors of the following: pUC series (Fermentas Life Sciences), pBluescript series (Stratagene, lahough, california), pET series (Novagen, madison, wis.), pGEX series (Pharmacia Biotech, uppsala, sweden) or pEX series (Clontech, palo Altuo, california). In some cases, phage vectors such as λg10, λgt11, λ ZapII (Stratagene), λembl4, and λnm1149 may also be used. In some embodiments, plant expression vectors may be used and include pBI01, pBI101.2, pBI101.3, pBI121, and pBIN19 (Clontech). In some embodiments, animal expression vectors include pEUK-Cl, pMAM and pMAMneo (Clontech). In some embodiments, a viral vector, such as a retroviral vector, is used.
In some embodiments, standard recombinant DNA techniques may be used to prepare recombinant expression vectors. In some embodiments, the vector may contain regulatory sequences, such as transcription and translation initiation and termination codons, which are specific for the type of host (e.g., bacterial, fungal, plant, or animal) into which the vector is introduced, as appropriate and considering whether the vector is DNA-based or RNA-based. In some embodiments, the vector may contain a non-native promoter operably linked to a nucleotide sequence encoding a TCR or antigen-binding portion (or other MHC-peptide binding molecule). In some embodiments, the promoter may be a non-viral promoter or a viral promoter, such as the Cytomegalovirus (CMV) promoter, the SV40 promoter, the RSV promoter, and promoters found in the long terminal repeat of murine stem cell viruses. Other known promoters are also contemplated.
In some embodiments, to generate a vector encoding a TCR, the α and β chains are PCR amplified from total cDNA isolated from T cell clones expressing the TCR of interest and cloned into an expression vector. In some embodiments, the α and β chains are cloned into the same vector. In some embodiments, the α and β chains are cloned into different vectors. In some embodiments, the generated alpha and beta strands are incorporated into a retroviral (e.g., lentiviral) vector.
IV. methods of administration
Methods of use and uses of the compositions are also provided, such as for treating diseases, conditions, and disorders, e.g., cancer.
Methods of administering therapeutic cell compositions whose efficacy has been assessed according to the methods provided herein (e.g., section I), and the use of such cells, populations, and compositions for the treatment or prevention of diseases, conditions, and disorders (including cancer) are provided. Also provided are methods of use and uses of therapeutic cell compositions whose efficacy has been assessed according to the methods provided herein (e.g., section I), as well as uses of such therapeutic cell compositions for treating or preventing diseases, conditions, and disorders, including cancer. In particular embodiments, the cells, populations, and compositions are as those generated and engineered according to any of the provided methods. In some embodiments, the cells, populations, and compositions are administered to a subject or patient suffering from a particular disease or disorder to be treated, e.g., via adoptive cell therapy (e.g., adoptive T cell therapy). In some embodiments, cells and compositions prepared by the provided methods (e.g., engineered compositions and end-of-production compositions after incubation and/or other processing steps) are administered to a subject, such as a subject having or at risk of a disease or disorder. In some aspects, the methods thereby treat a disease or disorder (e.g., ameliorate one or more symptoms thereof), such as by alleviating tumor burden in a cancer that expresses an antigen recognized by an engineered T cell.
Such methods and uses include therapeutic methods and uses, for example, which involve administering cells and compositions prepared by the provided methods (e.g., engineered compositions after incubation and/or other processing steps and compositions at the end of production) to a subject suffering from a disease, condition, or disorder (e.g., cancer) to effect treatment of the disease or disorder. In some embodiments, the efficacy of the composition is determined according to the methods provided herein. Uses include the use of compositions in such methods and treatments, and the use of such compositions in the manufacture of medicaments for performing such methods of treatment. In some embodiments, the methods and uses thereby treat a disease or condition or disorder, such as a tumor or cancer, in a subject.
Methods of administration of cells for adoptive cell therapy are known and may be used in combination with the provided methods and compositions. For example, adoptive T cell therapy methods are described in, for example, U.S. patent application publication No. 2003/0170238 to grenberg et al; U.S. Pat. No. 4,690,915 to Rosenberg; rosenberg (2011) Nat Rev Clin Oncol.8 (10): 577-85). See, e.g., themeli et al (2013) Nat Biotechnol.31 (10): 928-933; tsukahara et al (2013) Biochem Biophys Res Commun 438 (1): 84-9; davila et al (2013) PLoS ONE 8 (4): e61338.
As used herein, a "subject" is a mammal, such as a human or other animal, and is typically a human. In some embodiments, the subject (e.g., patient) to whom the cell, population of cells, or composition is administered is a mammal, typically a primate, such as a human. In some embodiments, the primate is a monkey or ape. The subject may be male or female, and may be of any suitable age, including infant, juvenile, adolescent, adult and geriatric subjects. In some embodiments, the subject is a non-primate mammal, such as a rodent.
As used herein, "treatment" (and grammatical variants thereof such as "treatment") refers to the complete or partial improvement or alleviation of a disease or condition or disorder, or a symptom, adverse effect, or outcome or phenotype associated therewith. Desirable therapeutic effects include, but are not limited to, preventing the occurrence or recurrence of a disease, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease, preventing metastasis, decreasing the rate of disease progression, amelioration or palliation of the disease state, and remission or improved prognosis. The term does not imply a complete cure of the disease or complete elimination of any symptoms or effects on all symptoms or outcomes.
As used herein, "delay of progression of a disease" means delay, impediment, slowing, delay, stabilization, inhibition, and/or delay of progression of a disease (e.g., cancer). This delay may have varying lengths of time, depending on the medical history and/or the individual being treated. It will be apparent to those skilled in the art that a sufficient or significant delay may actually cover prophylaxis, as the individual will not suffer from the disease. For example, advanced cancers, such as metastasis, may be delayed in their progression.
As used herein, "preventing" includes providing prophylaxis with respect to the occurrence or recurrence of a disease in a subject who may be susceptible to the disease but who has not yet been diagnosed with the disease. In some embodiments, the provided cells and compositions are used to delay the progression of a disease or delay the progression of a disease.
As used herein, "inhibiting" a function or activity is reducing the function or activity when compared to otherwise identical conditions other than the target condition or parameter, or alternatively, when compared to another instance. For example, cells that inhibit tumor growth reduce the growth rate of a tumor compared to the growth rate of a tumor in the absence of the cells.
In the context of administration, an "effective amount" of an agent (e.g., a pharmaceutical formulation, cell, or composition) refers to an amount effective to achieve a desired result (e.g., a therapeutic or prophylactic result) at the requisite dose/amount and for the requisite period of time.
A "therapeutically effective amount" of an agent (e.g., a pharmaceutical formulation or cell) refers to an amount effective to achieve a desired therapeutic result (e.g., treatment for a disease, condition, or disorder) and/or a pharmacokinetic or pharmacodynamic effect of the treatment at the desired dose and for the desired period of time. The therapeutically effective amount can vary depending on factors such as: disease state, age, sex and weight of the subject and the cell population administered. In some embodiments, provided methods involve administering cells and/or compositions in an effective amount (e.g., a therapeutically effective amount). In some embodiments, the determined efficacy of the therapeutic cell composition is used to determine an effective amount, e.g., a therapeutically effective amount.
"prophylactically effective amount" means an amount effective to achieve the desired prophylactic result at the requisite dosage and for the requisite period of time. Typically, but not necessarily, since the prophylactic dose is administered in the subject prior to or early in the disease, the prophylactically effective amount will be less than the therapeutically effective amount. In some embodiments, the determined efficacy of the therapeutic cell composition is used to determine a prophylactically effective amount.
The disease or condition being treated may be any disease or condition in which expression of the antigen is associated with and/or involved in the etiology of the disease, condition or disorder, e.g., causes, exacerbates or otherwise participates in such disease, condition or disorder. Exemplary diseases and conditions may include diseases or conditions associated with malignancy or cellular transformation (e.g., cancer), autoimmune or inflammatory diseases, or infectious diseases caused, for example, by bacteria, viruses, or other pathogens. Exemplary antigens, including antigens associated with various diseases and conditions that may be treated, are described above. In certain embodiments, the chimeric antigen receptor or transgenic TCR specifically binds to an antigen associated with a disease or disorder.
Thus, the methods and uses provided include methods and uses for adoptive cell therapy. In some embodiments, the method comprises administering a cell or a composition comprising the cell to a subject, tissue, or cell, e.g., a subject, tissue, or cell having, at risk of having, or suspected of having a disease, condition, or disorder. In some embodiments, the cells, populations, and compositions are administered to a subject having a particular disease or disorder to be treated, for example, by adoptive cell therapy, such as adoptive T cell therapy. In some embodiments, administration of the cell or composition to a subject (e.g., a subject suffering from or at risk of suffering from the disease or disorder) ameliorates one or more symptoms of the disease or disorder.
In some embodiments, cell therapy (e.g., adoptive T cell therapy) is performed by autologous transfer, wherein cells are isolated and/or otherwise prepared from a subject receiving the cell therapy or from a sample derived from such a subject. Thus, in some aspects, the cells are derived from a subject (e.g., patient) in need of treatment, and the cells are administered to the same subject after isolation and processing.
In some embodiments, cell therapy (e.g., adoptive T cell therapy) is performed by allogeneic transfer, wherein the cells are isolated and/or otherwise prepared from a subject other than the subject (e.g., the first subject) that is about to receive or ultimately receive the cell therapy. In such embodiments, the cells are then administered to a different subject of the same species, e.g., a second subject. In some embodiments, the first subject and the second subject are genetically identical. In some embodiments, the first subject and the second subject are genetically similar. In some embodiments, the second subject expresses the same HLA class or supertype as the first subject. The cells may be administered by any suitable means. The administration and administration may depend in part on whether the administration is brief or chronic. Various dosing schedules include, but are not limited to, single or multiple administrations at different points in time, bolus administrations, and pulse infusion.
In certain embodiments, the cell or individual cell subtype population is administered to the subject in a range of about 100 to about 1000 million cells and/or the amount of cells per kilogram body weight, e.g., 100 to about 500 million cells (e.g., about 500 tens of thousands of cells, about 2500 tens of thousands of cells, about 5 hundreds of millions of cells, about 10 hundreds of millions of cells, about 50 hundreds of millions of cells, about 200 hundreds of millions of cells, about 300 hundreds of millions of cells, about 400 hundreds of cells, or a range defined by any two of the foregoing values), such as about 1000 tens of thousands to about 1000 hundreds of millions of cells (e.g., about 2000 ten thousand cells, about 3000 ten thousand cells, about 4000 ten thousand cells, about 6000 ten thousand cells, about 7000 ten thousand cells, about 8000 ten thousand cells, about 9000 ten thousand cells, about 100 hundred million cells, about 250 hundred million cells, about 500 hundred million cells, about 750 hundred million cells, about 900 hundred million cells, or a range defined by any two of the foregoing values), and in some cases about 1 hundred million cells to about 500 hundred million cells (e.g., about 1.2 hundred million cells, about 2.5 hundred million cells, about 3.5 hundred million cells, about 4.5 hundred million cells, about 6.5 hundred million cells, about 8 hundred million cells, about 9 hundred million cells, about 30 hundred million cells, about 300 hundred million cells, about 450 hundred million cells), or any value in between these ranges and/or these ranges per kilogram of body weight. Likewise, the dosage may vary depending on the disease or disorder and/or the patient and/or other treatment-specific attributes. In some embodiments, the dosage may depend on the efficacy of the therapeutic cell composition. In some embodiments, the cells are administered as part of a combination therapy, such as simultaneously with another therapeutic intervention, such as an antibody or engineered cell or receptor or agent (e.g., a cytotoxic or therapeutic agent), or sequentially in any order. In some embodiments, the cells are co-administered with one or more additional therapeutic agents or administered in combination with another therapeutic intervention (administered simultaneously or sequentially in any order). In some cases, the cells are co-administered with another therapy in sufficiently close temporal proximity that the population of cells enhances the effect of one or more additional therapeutic agents, or vice versa. In some embodiments, the cells are administered prior to the one or more additional therapeutic agents. In some embodiments, the cells are administered after one or more additional therapeutic agents. In some embodiments, the one or more additional agents include a cytokine (e.g., IL-2), e.g., to enhance persistence. In some embodiments, the method comprises administering a chemotherapeutic agent.
In some embodiments, the biological activity of the engineered cell population (e.g., therapeutic cell composition) is measured after administration of the cells, for example, by any of a number of known methods. Parameters to be assessed include specific binding of engineered or natural T cells or other immune cells to an antigen, which is assessed in vivo, e.g., by imaging, or ex vivo, e.g., by ELISA or flow cytometry. In certain embodiments, the ability of an engineered cell to destroy a target cell can be measured using any suitable method known in the art, such as cytotoxicity assays described, for example, in: kochenderfer et al, J.Immunotherapy,32 (7): 689-702 (2009), and Herman et al, J.Immunogically Methods,285 (1): 25-40 (2004). In certain embodiments, the biological activity of the cells is measured by assaying the expression and/or secretion of one or more cytokines (e.g., CD 107a, IFNγ, IL-2, and TNF). In some aspects, biological activity is measured by assessing clinical outcome (e.g., reduction in tumor burden or burden).
In certain embodiments, the engineered cells are further modified in any number of ways such that their therapeutic or prophylactic efficacy is increased. For example, a population expressed engineered CAR or TCR can be conjugated directly or indirectly through a linker to a targeting moiety. Practices for conjugating a compound (e.g., CAR or TCR) to a targeting moiety are known in the art. See, e.g., wadwa et al, J.drug Targeting 3:1 1 1 (1995) and U.S. Pat. No. 5,087,616. In some embodiments, confirmation of increased therapeutic or prophylactic efficacy is determined using the methods of assessing efficacy described herein (e.g., section I).
Definition of IV
Unless defined otherwise, all technical, symbolic, and other technical and scientific terms or words used herein are intended to have the same meaning as commonly understood by one of ordinary skill in the art to which claimed subject matter pertains. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ease of reference, and such definitions contained herein should not be construed as representing substantial differences from the commonly understood meaning in the art.
As used herein, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. For example, "a" or "an" means "at least one" or "one or more". It is to be understood that the aspects and variations described herein include "consisting of" and/or "consisting essentially of" the aspects and variations.
Throughout this disclosure, various aspects of the claimed subject matter are presented in a range format. It should be understood that the description of the range format is merely for convenience and brevity and should not be interpreted as a inflexible limitation on the scope of the claimed subject matter. Accordingly, the description of a range should be considered to specifically disclose all possible sub-ranges as well as individual values within the range. For example, where a range of values is provided, it is to be understood that each intervening value, between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the claimed subject matter. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the claimed subject matter, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the claimed subject matter. This applies regardless of the width of the range.
The term "about" as used herein refers to a common error range for the corresponding values as readily known to those skilled in the art. References herein to "about" a value or parameter include (and describe) implementations directed to the value or parameter itself. For example, a description referring to "about X" includes a description of "X".
As used herein, recitation of a nucleotide or amino acid position "corresponding to" a nucleotide or amino acid position in a disclosed sequence (as shown in the sequence listing) refers to the identified nucleotide or amino acid position after alignment with the disclosed sequence using a standard alignment algorithm (e.g., the GAP algorithm) to maximize identity. By aligning the sequences, the person skilled in the art can, for example, use conserved and identical amino acid residues as guidance to identify the corresponding residues. Typically, to identify the corresponding positions, the amino acid sequences are aligned such that a highest order match is obtained (see, e.g., computational Molecular Biology, lesk, a.m. edit, oxford University Press, new York,1988;Biocomputing:Informatics and Genome Projects,Smith,D.W. Edit, academic Press, new York,1993;Computer Analysis ofSequence Data,Part I,Griffin,A.M. And Griffin, h.g. edit, humana Press, new Jersey,1994;Sequence Analysis in Molecular Biology,von Heinje,G., academic Press,1987; and Sequence Analysis Primer, gribskov, m. and deveux, j. Edit, M Stockton Press, new York,1991; carrillo et al (1988) SIAM J Applied Math 48:1073).
As used herein, the term "vector" refers to a nucleic acid molecule capable of transmitting another nucleic acid to which it is linked. The term includes vectors that are self-replicating nucleic acid structures and that incorporate into the genome of a host cell into which they have been introduced. Certain vectors are capable of directing the expression of nucleic acids to which they are operatively linked. Such vectors are referred to herein as "expression vectors". Vectors include viral vectors such as retroviral (e.g., gamma-retrovirus and lentivirus) vectors.
The terms "host cell", "host cell line", and "host cell culture" are used interchangeably and refer to cells into which exogenous nucleic acid has been introduced, including the progeny of such cells. Host cells include "transformants" and "transformed cells" which include primary transformed cells and progeny derived therefrom, irrespective of the number of passages. The nucleic acid content of the offspring may not be exactly the same as the parent cell, but may contain mutations. Included herein are mutant progeny that have the same function or biological activity as selected or selected in the original transformed cell.
As used herein, a statement that a cell or cell population is "positive" for a particular marker refers to the detectable presence of the particular marker (typically a surface marker) on or in the cell. When referring to a surface marker, the term refers to the presence of surface expression as detected by flow cytometry, e.g., by staining with an antibody that specifically binds to the marker and detecting the antibody, wherein the staining is detectable by flow cytometry at a level that is substantially higher than that detected by the same procedure with an isotype-matched control under otherwise identical conditions, and/or that is substantially similar to that of cells known to be positive for the marker, and/or that is substantially higher than that of cells known to be negative for the marker.
As used herein, a statement that a cell or cell population is "negative" for a particular marker means that the particular marker (typically a surface marker) is not present on or in the cell in a substantially detectable manner. When referring to a surface marker, the term refers to the absence of surface expression as detected by flow cytometry, e.g., by staining with an antibody that specifically binds to the marker and detecting the antibody, wherein the staining is not detected by flow cytometry at a level that is substantially higher than that detected by the same procedure with an isotype-matched control under otherwise identical conditions, and/or that is substantially lower than that of cells known to be positive for the marker, and/or that is substantially similar to that of cells known to be negative for the marker.
As used herein, "percent amino acid sequence identity (%)" and "percent identity" when used with respect to an amino acid sequence (reference polypeptide sequence) are defined as the percentage of amino acid residues in a candidate sequence (e.g., a subject antibody or fragment) that are identical to amino acid residues in the reference polypeptide sequence after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity and not considering any conservative substitutions as part of the sequence identity. Alignment for the purpose of determining the percent amino acid sequence identity may be accomplished in a variety of ways well known in the art, for example, using publicly available computer software such as BLAST, BLAST-2, ALIGN or Megalign (DNASTAR) software. One skilled in the art can determine the appropriate parameters for aligning sequences, including any algorithms needed to achieve maximum alignment over the full length of the sequences compared.
Amino acid substitutions may include substitution of one amino acid for another amino acid in the polypeptide. Substitutions may be conservative amino acid substitutions or non-conservative amino acid substitutions. Amino acid substitutions may be introduced into binding molecules of interest (e.g., antibodies), and products screened for a desired activity (e.g., retention/improved antigen binding, reduced immunogenicity, or improved ADCC or CDC).
Amino acids can generally be grouped according to the following common side chain characteristics:
(1) Hydrophobicity: norleucine, met, ala, val, leu, ile;
(2) Neutral hydrophilicity: cys, ser, thr, asn, gln;
(3) Acid: asp, glu;
(4) Alkaline: his, lys, arg;
(5) Residues that affect chain orientation: gly, pro;
(6) Aromatic: trp, tyr, phe.
In some embodiments, conservative substitutions may involve replacing a member of one of these classes with another member of the same class. In some embodiments, non-conservative amino acid substitutions may involve exchanging members of one of these classes for another class.
As used herein, a composition refers to any mixture of two or more products, substances, or compounds (including cells). It may be a solution, suspension, liquid, powder, paste, aqueous, non-aqueous, or any combination thereof.
As used herein, a "subject" is a mammal, such as a human or other animal, and is typically a human.
Unless defined otherwise, all technical, symbolic, and other technical and scientific terms or words used herein are intended to have the same meaning as commonly understood by one of ordinary skill in the art to which claimed subject matter pertains. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ease of reference, and such definitions contained herein should not be construed as representing substantial differences from the commonly understood meaning in the art.
V. exemplary embodiments
The provided embodiments include:
1. a method of determining the efficacy of a therapeutic cell composition, the method comprising:
performing a plurality of incubations, each of the plurality of incubations comprising incubating cells of a therapeutic cell composition comprising cells engineered to express a recombinant receptor with a recombinant receptor stimulator, wherein:
binding of the recombinant receptor stimulator to the recombinant receptor stimulates recombinant receptor-dependent activity of the cell; and is also provided with
Each of the plurality of incubations includes a different stepwise adjustment ratio of cells of the therapeutic cell composition to the recombinant receptor stimulator;
Measuring recombinant receptor-dependent activity from each of the plurality of incubations;
based on the recombinant receptor-dependent activity measured from each of the plurality of incubations, a stepwise adjustment ratio is determined that results in a half-maximal recombinant receptor-dependent activity.
2. The method of embodiment 1, further comprising determining the relative efficacy of the therapeutic cell composition by comparing the stepwise adjustment ratio of the half-maximal recombinant receptor-dependent activity resulting in the therapeutic cell composition to the stepwise adjustment ratio of the half-maximal recombinant receptor-dependent activity resulting in a reference standard.
3. A method of determining the efficacy of a therapeutic cell composition, the method comprising:
performing a plurality of incubations, each of the plurality of incubations comprising incubating cells of a therapeutic cell composition comprising cells engineered to express a recombinant receptor with a recombinant receptor stimulator, wherein:
binding of the recombinant receptor stimulator to the recombinant receptor stimulates recombinant receptor-dependent activity of the cell; and is also provided with
Each of the plurality of incubations includes a different stepwise adjustment ratio of cells of the therapeutic cell composition to the recombinant receptor stimulator;
Measuring recombinant receptor-dependent activity from each of the plurality of incubations; and
the relative potency of the therapeutic cell composition is determined by comparing the half maximal recombinant receptor-dependent activity of the therapeutic cell composition to the half maximal recombinant receptor-dependent activity of a reference standard.
4. The method of any one of embodiments 1-3, wherein each of the plurality of incubations comprises culturing a constant number of cells of the resulting therapeutic composition with different amounts of the recombinant receptor stimulator to a plurality of different stepwise adjustment ratios.
5. The method of any one of embodiments 1-3, wherein each of the plurality of incubations comprises culturing a constant amount of binding molecule with a different number of cells of the therapeutic composition to generate a plurality of different stepwise adjustment ratios.
6. The method of any one of embodiments 1-5, wherein the plurality of incubations is performed on two or more, optionally 3, 4, 5, 6, 7, 8, 9, 10 or more therapeutic cell compositions.
7. The method of embodiment 6, wherein the two or more therapeutic cell compositions each comprise the same recombinant receptor.
8. The method of embodiment 6, wherein the two or more therapeutic cell compositions each comprise a different recombinant receptor.
9. The method of embodiment 6, wherein at least one of the two or more therapeutic cell compositions comprises a recombinant receptor that is different from the other therapeutic compositions.
10. The method of any one of embodiments 6-9, wherein each of the two or more therapeutic cell compositions are manufactured using the same manufacturing process.
11. The method of any one of embodiments 6-9, wherein the two or more therapeutic cell compositions are each manufactured using a different manufacturing process.
12. The method of any one of embodiments 6-9, wherein at least one of the two or more therapeutic cell compositions is manufactured using a manufacturing process that is different from the manufacturing process used to manufacture the other therapeutic cell compositions.
13. The method of any one of embodiments 6-12, wherein the two or more therapeutic cell compositions are produced by cells from a single subject.
14. The method of any one of embodiments 6-12, wherein the two or more therapeutic cell compositions are produced by cells from different subjects.
15. The method of embodiment 13 or embodiment 14, wherein the subject is a healthy subject or a subject with a disease or disorder.
16. The method of any one of embodiments 1-15, wherein the plurality of incubations is at least three incubations.
17. The method of any one of embodiments 1-16, wherein the plurality of incubations is at least five incubations.
18. The method of any one of embodiments 1-17, wherein the plurality of incubations is at least seven incubations.
19. The method of any one of embodiments 1-18, wherein the plurality of incubations is at least ten incubations.
20. The method of any one of embodiments 1-19, wherein the recombinant receptor-dependent activity comprises one or more of: cytokine expression, cytolytic activity, receptor up-regulation, receptor down-regulation, proliferation, gene up-regulation, gene down-regulation, or cellular health.
21. The method of any one of embodiments 1-20, wherein the recombinant receptor-dependent activity comprises or is cytokine expression or production.
22. The method of any one of embodiments 1-21, wherein the recombinant receptor-dependent activity comprises or is cytokine expression or production, wherein the cytokine is TNF- α, IFN- γ (IFNg), or IL-2.
23. The method of any one of embodiments 1-22, wherein the recombinant receptor-dependent activity comprises or is cytolytic activity.
24. The method of any one of embodiments 1-23, wherein the recombinant receptor-dependent activity comprises or is receptor up-regulation.
25. The method of any one of embodiments 1-24, wherein the recombinant receptor-dependent activity comprises or is receptor down-regulation.
26. The method of any one of embodiments 1-25, wherein the recombinant receptor-dependent activity comprises or is proliferation.
27. The method of any one of embodiments 1-26, wherein the recombinant receptor-dependent activity comprises or is gene up-regulation.
28. The method of any one of embodiments 1-27, wherein the recombinant receptor-dependent activity comprises or is down-regulated.
29. The method of any one of embodiments 1-28, wherein the recombinant receptor-dependent activity comprises or is cellular health.
30. The method of any one of embodiments 1-29, wherein the recombinant receptor-dependent activity comprises or is a cellular health, wherein the cellular health comprises one or more of cell death, cell diameter, viable cell concentration, and cell count.
31. The method of any one of embodiments 1-30, wherein the recombinant receptor-dependent activity measured at each of the plurality of incubations is normalized to a maximum receptor-dependent activity measured for the therapeutic cell composition.
32. The method of any one of embodiments 1-31, wherein the reference standard is a therapeutic cell composition comprising a validated step-by-step adjustment ratio that results in half-maximal recombinant receptor-dependent activity, a commercially available therapeutic cell composition, a therapeutic cell composition manufactured using the same manufacturing process as that used to manufacture the therapeutic cell composition, a therapeutic cell composition manufactured using a different manufacturing process than that used to manufacture the therapeutic cell composition, a therapeutic cell composition comprising the same recombinant receptor as that therapeutic cell composition, a therapeutic cell composition comprising a different recombinant receptor than that therapeutic cell composition, a therapeutic cell composition manufactured from the same subject, or a therapeutic cell composition manufactured from a different subject.
33. The method of any one of embodiments 6-32, wherein the reference standard is one of the two or more therapeutic compositions.
34. The method of any one of embodiments 1-33, wherein the recombinant receptor stimulant comprises a target antigen of the recombinant receptor or an extracellular domain-binding portion thereof, optionally a recombinant antigen.
35. The method of embodiment 34, wherein the recombinant receptor stimulant comprises an ectodomain binding portion of the antigen, and the ectodomain binding portion comprises an epitope recognized by the recombinant receptor.
36. The method of any one of embodiments 1-33, wherein the recombinant receptor stimulant is an anti-idiotype antibody specific for an extracellular antigen-binding domain of the recombinant receptor.
37. The method of any one of embodiments 1-36, wherein the recombinant receptor stimulant is immobilized or attached to a solid support.
38. The method of embodiment 37, wherein the solid support is a surface of a vessel, optionally a well of a microplate, in which the plurality of incubations are performed.
39. The method of embodiment 37, wherein the solid support is a bead.
40. The method of any one of embodiments 1-33, wherein the recombinant receptor stimulant is an antigen-expressing cell, optionally wherein the cell is a clone, derived from a cell line, or a primary cell taken from a subject.
41. The method of embodiment 40, wherein the antigen expressing cell is a cell line.
42. The method of embodiment 41, wherein the cell line is a tumor cell line.
43. The method of embodiment 40, wherein the antigen expressing cell is a cell that has been introduced, optionally by transduction, to express an antigen of the recombinant receptor.
44. The method of any one of embodiments 1-43, wherein the stepwise adjustment ratio reaches a linear dose response range of recombinant receptor-dependent activity of the reference standard.
45. The method of embodiment 44, wherein the stepwise adjustment ratio comprises a lower asymptote (minimum) and a higher asymptote (maximum) recombinant receptor-dependent activity of the reference standard.
46. The method of any one of embodiments 1-35, wherein the therapeutic cell composition comprises a single cell subtype enriched or purified from a biological sample or a mixed population of cell subtypes obtained optionally by mixing cell subtypes enriched or purified from a biological sample.
47. The method of embodiment 46, wherein the biological sample comprises a whole blood sample, a buffy coat sample, a Peripheral Blood Mononuclear Cell (PBMC) sample, an unfractionated cell sample, a lymphocyte sample, a leukocyte sample, a apheresis product, or a leukocyte apheresis product.
48. The method of any one of embodiments 1-47, wherein the therapeutic cell composition comprises primary cells.
49. The method of any one of embodiments 1-38, wherein the therapeutic cell composition comprises autologous cells from the subject to be treated.
50. The method of any one of embodiments 1-49, wherein the therapeutic cell composition comprises allogeneic cells.
51. The method of any one of embodiments 1-50, wherein the therapeutic cell composition comprises cd3+, cd4+, and/or cd8+ T cells.
52. The method of any one of embodiments 1-51, wherein the therapeutic cell composition comprises or is a cd4+ T cell.
53. The method of any one of embodiments 1-52, wherein the therapeutic cell composition comprises or is a cd8+ T cell.
54. The method of any one of embodiments 1-53, wherein the recombinant receptor is a Chimeric Antigen Receptor (CAR).
55. The method of any one of embodiments 1-54, wherein the plurality of incubations are performed in flasks, tubes, or multi-well plates.
56. The method of any one of embodiments 1-55, wherein each of the plurality of incubations is performed separately in a well of a multi-well plate.
57. The method of embodiment 55 or embodiment 56, wherein the multi-well plate is a 96-well plate, a 48-well plate, a 12-well plate, or a 6-well plate.
58. The method of any one of embodiments 1 and 4-57, further comprising determining a dose of cells of the therapeutic composition for administration to a subject in need thereof based on a stepwise adjustment ratio that results in half-maximal recombinant receptor-dependent activity.
59. The method of any one of embodiments 2-57, further comprising determining a dose of cells of the therapeutic composition for administration to a subject in need thereof based on the relative efficacy.
60. The method of embodiment 58 or embodiment 59, wherein the subject has a disease or disorder.
61. The method of embodiment 60, wherein the disease or disorder is cancer.
62. The method of any one of embodiments 2-61, further comprising determining a manufacturing process that produces an optimal therapeutic cell composition efficacy based on the relative efficacy, wherein the optimal therapeutic cell composition efficacy is associated with a complete and/or sustained response and/or reduced toxicity.
63. The method of any one of embodiments 2-62, further comprising determining a manufacturing process that produces a therapeutic cell composition having a reduced or low difference in potency based on the relative potency, wherein the reduced or low difference is determined by comparison with a difference in a different manufacturing process.
VI. Examples
The following examples are included for illustrative purposes only and are not intended to limit the scope of the invention.
Example 1: an assay for assessing the sensitivity of a cell therapy product to an antigen stimulus.
The assay is designed to measure the sensitivity of therapeutic cell compositions containing cells expressing recombinant receptors (e.g., chimeric antigen receptors) to antigen stimuli.
Primary cd4+ and cd8+ T cells from three patients or healthy human donors were selected from isolated PBMCs from donor leukocyte apheresis samples. CD4+ and CD8+ T cells are stimulated in serum-free medium in the presence of recombinant IL-2, IL-7 and IL-15, in the presence of anti-CD 3 and anti-CD 28 antibodies or binding fragments. Stimulation was performed between 18 and 30 hours by incubation. Cells are transduced with lentiviral vectors encoding Chimeric Antigen Receptors (CARs) against a specific antigen (e.g., CD19 or BCMA). The CAR contains an scFv antigen binding domain specific for an antigen (e.g., CD19 or BCMA), an immunoglobulin spacer, a transmembrane domain (e.g., from human CD 28), and an intracellular signaling domain containing a human CD 3-zeta intracellular signaling domain and a costimulatory signaling domain (e.g., a human 4-1BB intracellular signaling domain). The transduced cells are then incubated in the presence of recombinant cytokines for expansion. Although this example is illustrated with CAR-expressing T cells generated, this assay can be used to assess the antigen-specific sensitivity of any antigen-directed recombinant receptor.
To measure the sensitivity of exemplary therapeutic cell compositions to antigen-specific stimuli, a fixed concentration of therapeutic cell composition is incubated with a stepwise adjusted amount of antigen-expressing target cells. About 50,000 car+ T cells of the therapeutic cell composition (which act as effector cells in this assay) were added to each well of the multi-well plate. Antigen expressing target cells were added in stepwise adjustment of antigen expressing target cell to effector cell ratio (T: E ratio) from 12:1 to 0.012:1. Cells were co-cultured for between 2 and 48 hours, and then antigen-specific responses were assessed by monitoring the functional activity of T cells. In this exemplary assay, supernatants were collected after 16 hours to assess antigen-specific cytokine production.
FIG. 1A shows exemplary secreted cytokine IFNγ concentrations at each T:E ratio for donors according to three exemplary generated cell products. Fig. 1B shows cytokine secretion normalized to the maximum cytokine concentration observed at the higher asymptote (Vmax) (y-axis).
The ratio of T to E for each donor producing 50% cytokine secretion (e.g., 50% effective stimulation or ES 50) was determined and relative potency was calculated relative to donor 1 (reference standard). For comparison, three different cell composition products were evaluated using a traditional assay format in which cytokine secretion under maximum antigen stimulation of the drug product was measured and relative potency was also calculated relative to donor 1. The results are shown in Table E1. The results described in table E1 show the different relationship between the amount of cytokines secreted by cells of the therapeutic cell composition at maximum antigen-specific stimulation and their sensitivity to antigen-specific stimulation, as determined by the ratio of T: E at 50% effective stimulation (ES 50). These results indicate that conventional assays may result in saturation of antigen stimulation levels, which may not provide an accurate measure of antigen sensitivity to antigen stimulation, while the provided assays may more reliably measure the sensitivity of antigen-directed therapeutic cell compositions to antigen stimulation.
Table e1. Relative efficacy as determined by the assay.
The present invention is not intended to be limited in scope by the specific disclosed embodiments, which are provided, for example, to illustrate various aspects of the invention. Various modifications to the compositions and methods will be apparent from the description and teachings herein. Such changes may be practiced without departing from the true scope and spirit of the disclosure, and are intended to fall within the scope of the disclosure.
Sequence(s)
/>
/>
/>
/>
/>
/>
Sequence listing
<110> Cino therapeutics Co., ltd
<120> method of determining efficacy of therapeutic cell compositions
<130> 73504-20230.40
<140> not yet allocated
<141> along with the submission
<150> 63/164,527
<151> 2021-03-22
<160> 92
<170> FastSEQ version 4.0 for Windows
<210> 1
<211> 24
<212> PRT
<213> artificial sequence
<220>
<223> T2A
<400> 1
Leu Glu Gly Gly Gly Glu Gly Arg Gly Ser Leu Leu Thr Cys Gly Asp
1 5 10 15
Val Glu Glu Asn Pro Gly Pro Arg
20
<210> 2
<211> 357
<212> PRT
<213> artificial sequence
<220>
<223> tEGFR
<400> 2
Met Leu Leu Leu Val Thr Ser Leu Leu Leu Cys Glu Leu Pro His Pro
1 5 10 15
Ala Phe Leu Leu Ile Pro Arg Lys Val Cys Asn Gly Ile Gly Ile Gly
20 25 30
Glu Phe Lys Asp Ser Leu Ser Ile Asn Ala Thr Asn Ile Lys His Phe
35 40 45
Lys Asn Cys Thr Ser Ile Ser Gly Asp Leu His Ile Leu Pro Val Ala
50 55 60
Phe Arg Gly Asp Ser Phe Thr His Thr Pro Pro Leu Asp Pro Gln Glu
65 70 75 80
Leu Asp Ile Leu Lys Thr Val Lys Glu Ile Thr Gly Phe Leu Leu Ile
85 90 95
Gln Ala Trp Pro Glu Asn Arg Thr Asp Leu His Ala Phe Glu Asn Leu
100 105 110
Glu Ile Ile Arg Gly Arg Thr Lys Gln His Gly Gln Phe Ser Leu Ala
115 120 125
Val Val Ser Leu Asn Ile Thr Ser Leu Gly Leu Arg Ser Leu Lys Glu
130 135 140
Ile Ser Asp Gly Asp Val Ile Ile Ser Gly Asn Lys Asn Leu Cys Tyr
145 150 155 160
Ala Asn Thr Ile Asn Trp Lys Lys Leu Phe Gly Thr Ser Gly Gln Lys
165 170 175
Thr Lys Ile Ile Ser Asn Arg Gly Glu Asn Ser Cys Lys Ala Thr Gly
180 185 190
Gln Val Cys His Ala Leu Cys Ser Pro Glu Gly Cys Trp Gly Pro Glu
195 200 205
Pro Arg Asp Cys Val Ser Cys Arg Asn Val Ser Arg Gly Arg Glu Cys
210 215 220
Val Asp Lys Cys Asn Leu Leu Glu Gly Glu Pro Arg Glu Phe Val Glu
225 230 235 240
Asn Ser Glu Cys Ile Gln Cys His Pro Glu Cys Leu Pro Gln Ala Met
245 250 255
Asn Ile Thr Cys Thr Gly Arg Gly Pro Asp Asn Cys Ile Gln Cys Ala
260 265 270
His Tyr Ile Asp Gly Pro His Cys Val Lys Thr Cys Pro Ala Gly Val
275 280 285
Met Gly Glu Asn Asn Thr Leu Val Trp Lys Tyr Ala Asp Ala Gly His
290 295 300
Val Cys His Leu Cys His Pro Asn Cys Thr Tyr Gly Cys Thr Gly Pro
305 310 315 320
Gly Leu Glu Gly Cys Pro Thr Asn Gly Pro Lys Ile Pro Ser Ile Ala
325 330 335
Thr Gly Met Val Gly Ala Leu Leu Leu Leu Leu Val Val Ala Leu Gly
340 345 350
Ile Gly Leu Phe Met
355
<210> 3
<211> 335
<212> PRT
<213> artificial sequence
<220>
<223> tEGFR
<400> 3
Arg Lys Val Cys Asn Gly Ile Gly Ile Gly Glu Phe Lys Asp Ser Leu
1 5 10 15
Ser Ile Asn Ala Thr Asn Ile Lys His Phe Lys Asn Cys Thr Ser Ile
20 25 30
Ser Gly Asp Leu His Ile Leu Pro Val Ala Phe Arg Gly Asp Ser Phe
35 40 45
Thr His Thr Pro Pro Leu Asp Pro Gln Glu Leu Asp Ile Leu Lys Thr
50 55 60
Val Lys Glu Ile Thr Gly Phe Leu Leu Ile Gln Ala Trp Pro Glu Asn
65 70 75 80
Arg Thr Asp Leu His Ala Phe Glu Asn Leu Glu Ile Ile Arg Gly Arg
85 90 95
Thr Lys Gln His Gly Gln Phe Ser Leu Ala Val Val Ser Leu Asn Ile
100 105 110
Thr Ser Leu Gly Leu Arg Ser Leu Lys Glu Ile Ser Asp Gly Asp Val
115 120 125
Ile Ile Ser Gly Asn Lys Asn Leu Cys Tyr Ala Asn Thr Ile Asn Trp
130 135 140
Lys Lys Leu Phe Gly Thr Ser Gly Gln Lys Thr Lys Ile Ile Ser Asn
145 150 155 160
Arg Gly Glu Asn Ser Cys Lys Ala Thr Gly Gln Val Cys His Ala Leu
165 170 175
Cys Ser Pro Glu Gly Cys Trp Gly Pro Glu Pro Arg Asp Cys Val Ser
180 185 190
Cys Arg Asn Val Ser Arg Gly Arg Glu Cys Val Asp Lys Cys Asn Leu
195 200 205
Leu Glu Gly Glu Pro Arg Glu Phe Val Glu Asn Ser Glu Cys Ile Gln
210 215 220
Cys His Pro Glu Cys Leu Pro Gln Ala Met Asn Ile Thr Cys Thr Gly
225 230 235 240
Arg Gly Pro Asp Asn Cys Ile Gln Cys Ala His Tyr Ile Asp Gly Pro
245 250 255
His Cys Val Lys Thr Cys Pro Ala Gly Val Met Gly Glu Asn Asn Thr
260 265 270
Leu Val Trp Lys Tyr Ala Asp Ala Gly His Val Cys His Leu Cys His
275 280 285
Pro Asn Cys Thr Tyr Gly Cys Thr Gly Pro Gly Leu Glu Gly Cys Pro
290 295 300
Thr Asn Gly Pro Lys Ile Pro Ser Ile Ala Thr Gly Met Val Gly Ala
305 310 315 320
Leu Leu Leu Leu Leu Val Val Ala Leu Gly Ile Gly Leu Phe Met
325 330 335
<210> 4
<211> 18
<212> PRT
<213> artificial sequence
<220>
<223> T2A
<400> 4
Glu Gly Arg Gly Ser Leu Leu Thr Cys Gly Asp Val Glu Glu Asn Pro
1 5 10 15
Gly Pro
<210> 5
<211> 22
<212> PRT
<213> artificial sequence
<220>
<223> P2A
<400> 5
Gly Ser Gly Ala Thr Asn Phe Ser Leu Leu Lys Gln Ala Gly Asp Val
1 5 10 15
Glu Glu Asn Pro Gly Pro
20
<210> 6
<211> 19
<212> PRT
<213> artificial sequence
<220>
<223> P2A
<400> 6
Ala Thr Asn Phe Ser Leu Leu Lys Gln Ala Gly Asp Val Glu Glu Asn
1 5 10 15
Pro Gly Pro
<210> 7
<211> 20
<212> PRT
<213> artificial sequence
<220>
<223> E2A
<400> 7
Gln Cys Thr Asn Tyr Ala Leu Leu Lys Leu Ala Gly Asp Val Glu Ser
1 5 10 15
Asn Pro Gly Pro
20
<210> 8
<211> 22
<212> PRT
<213> artificial sequence
<220>
<223> F2A
<400> 8
Val Lys Gln Thr Leu Asn Phe Asp Leu Leu Lys Leu Ala Gly Asp Val
1 5 10 15
Glu Ser Asn Pro Gly Pro
20
<210> 9
<211> 66
<212> DNA
<213> artificial sequence
<220>
<223> GMCSFR alpha chain signal sequence
<400> 9
atgcttctcc tggtgacaag ccttctgctc tgtgagttac cacacccagc attcctcctg 60
atccca 66
<210> 10
<211> 22
<212> PRT
<213> artificial sequence
<220>
<223> GMCSFR alpha chain signal sequence
<400> 10
Met Leu Leu Leu Val Thr Ser Leu Leu Leu Cys Glu Leu Pro His Pro
1 5 10 15
Ala Phe Leu Leu Ile Pro
20
<210> 11
<211> 18
<212> PRT
<213> artificial sequence
<220>
<223> CD8 alpha Signal peptide
<400> 11
Met Ala Leu Pro Val Thr Ala Leu Leu Leu Pro Leu Ala Leu Leu Leu
1 5 10 15
His Ala
<210> 12
<211> 16
<212> PRT
<213> artificial sequence
<220>
<223> CD33 Signal peptide
<400> 12
Met Pro Leu Leu Leu Leu Leu Pro Leu Leu Trp Ala Gly Ala Leu Ala
1 5 10 15
<210> 13
<211> 54
<212> PRT
<213> artificial sequence
<220>
<223> extracellular domain of human BCMA (GenBank accession number
NP_001183.2)
<400> 13
Met Leu Gln Met Ala Gly Gln Cys Ser Gln Asn Glu Tyr Phe Asp Ser
1 5 10 15
Leu Leu His Ala Cys Ile Pro Cys Gln Leu Arg Cys Ser Ser Asn Thr
20 25 30
Pro Pro Leu Thr Cys Gln Arg Tyr Cys Asn Ala Ser Val Thr Asn Ser
35 40 45
Val Lys Gly Thr Asn Ala
50
<210> 14
<211> 668
<212> PRT
<213> artificial sequence
<220>
<223> human CD22 extracellular Domain
<400> 14
Asp Ser Ser Lys Trp Val Phe Glu His Pro Glu Thr Leu Tyr Ala Trp
1 5 10 15
Glu Gly Ala Cys Val Trp Ile Pro Cys Thr Tyr Arg Ala Leu Asp Gly
20 25 30
Asp Leu Glu Ser Phe Ile Leu Phe His Asn Pro Glu Tyr Asn Lys Asn
35 40 45
Thr Ser Lys Phe Asp Gly Thr Arg Leu Tyr Glu Ser Thr Lys Asp Gly
50 55 60
Lys Val Pro Ser Glu Gln Lys Arg Val Gln Phe Leu Gly Asp Lys Asn
65 70 75 80
Lys Asn Cys Thr Leu Ser Ile His Pro Val His Leu Asn Asp Ser Gly
85 90 95
Gln Leu Gly Leu Arg Met Glu Ser Lys Thr Glu Lys Trp Met Glu Arg
100 105 110
Ile His Leu Asn Val Ser Glu Arg Pro Phe Pro Pro His Ile Gln Leu
115 120 125
Pro Pro Glu Ile Gln Glu Ser Gln Glu Val Thr Leu Thr Cys Leu Leu
130 135 140
Asn Phe Ser Cys Tyr Gly Tyr Pro Ile Gln Leu Gln Trp Leu Leu Glu
145 150 155 160
Gly Val Pro Met Arg Gln Ala Ala Val Thr Ser Thr Ser Leu Thr Ile
165 170 175
Lys Ser Val Phe Thr Arg Ser Glu Leu Lys Phe Ser Pro Gln Trp Ser
180 185 190
His His Gly Lys Ile Val Thr Cys Gln Leu Gln Asp Ala Asp Gly Lys
195 200 205
Phe Leu Ser Asn Asp Thr Val Gln Leu Asn Val Lys His Thr Pro Lys
210 215 220
Leu Glu Ile Lys Val Thr Pro Ser Asp Ala Ile Val Arg Glu Gly Asp
225 230 235 240
Ser Val Thr Met Thr Cys Glu Val Ser Ser Ser Asn Pro Glu Tyr Thr
245 250 255
Thr Val Ser Trp Leu Lys Asp Gly Thr Ser Leu Lys Lys Gln Asn Thr
260 265 270
Phe Thr Leu Asn Leu Arg Glu Val Thr Lys Asp Gln Ser Gly Lys Tyr
275 280 285
Cys Cys Gln Val Ser Asn Asp Val Gly Pro Gly Arg Ser Glu Glu Val
290 295 300
Phe Leu Gln Val Gln Tyr Ala Pro Glu Pro Ser Thr Val Gln Ile Leu
305 310 315 320
His Ser Pro Ala Val Glu Gly Ser Gln Val Glu Phe Leu Cys Met Ser
325 330 335
Leu Ala Asn Pro Leu Pro Thr Asn Tyr Thr Trp Tyr His Asn Gly Lys
340 345 350
Glu Met Gln Gly Arg Thr Glu Glu Lys Val His Ile Pro Lys Ile Leu
355 360 365
Pro Trp His Ala Gly Thr Tyr Ser Cys Val Ala Glu Asn Ile Leu Gly
370 375 380
Thr Gly Gln Arg Gly Pro Gly Ala Glu Leu Asp Val Gln Tyr Pro Pro
385 390 395 400
Lys Lys Val Thr Thr Val Ile Gln Asn Pro Met Pro Ile Arg Glu Gly
405 410 415
Asp Thr Val Thr Leu Ser Cys Asn Tyr Asn Ser Ser Asn Pro Ser Val
420 425 430
Thr Arg Tyr Glu Trp Lys Pro His Gly Ala Trp Glu Glu Pro Ser Leu
435 440 445
Gly Val Leu Lys Ile Gln Asn Val Gly Trp Asp Asn Thr Thr Ile Ala
450 455 460
Cys Ala Ala Cys Asn Ser Trp Cys Ser Trp Ala Ser Pro Val Ala Leu
465 470 475 480
Asn Val Gln Tyr Ala Pro Arg Asp Val Arg Val Arg Lys Ile Lys Pro
485 490 495
Leu Ser Glu Ile His Ser Gly Asn Ser Val Ser Leu Gln Cys Asp Phe
500 505 510
Ser Ser Ser His Pro Lys Glu Val Gln Phe Phe Trp Glu Lys Asn Gly
515 520 525
Arg Leu Leu Gly Lys Glu Ser Gln Leu Asn Phe Asp Ser Ile Ser Pro
530 535 540
Glu Asp Ala Gly Ser Tyr Ser Cys Trp Val Asn Asn Ser Ile Gly Gln
545 550 555 560
Thr Ala Ser Lys Ala Trp Thr Leu Glu Val Leu Tyr Ala Pro Arg Arg
565 570 575
Leu Arg Val Ser Met Ser Pro Gly Asp Gln Val Met Glu Gly Lys Ser
580 585 590
Ala Thr Leu Thr Cys Glu Ser Asp Ala Asn Pro Pro Val Ser His Tyr
595 600 605
Thr Trp Phe Asp Trp Asn Asn Gln Ser Leu Pro Tyr His Ser Gln Lys
610 615 620
Leu Arg Leu Glu Pro Val Lys Val Gln His Ser Gly Ala Tyr Trp Cys
625 630 635 640
Gln Gly Thr Asn Ser Val Gly Lys Gly Arg Ser Pro Leu Ser Thr Leu
645 650 655
Thr Val Tyr Tyr Ser Pro Glu Thr Ile Gly Arg Arg
660 665
<210> 15
<211> 556
<212> PRT
<213> Homo Sapiens (Homo Sapiens)
<220>
<223> CD19
<400> 15
Met Pro Pro Pro Arg Leu Leu Phe Phe Leu Leu Phe Leu Thr Pro Met
1 5 10 15
Glu Val Arg Pro Glu Glu Pro Leu Val Val Lys Val Glu Glu Gly Asp
20 25 30
Asn Ala Val Leu Gln Cys Leu Lys Gly Thr Ser Asp Gly Pro Thr Gln
35 40 45
Gln Leu Thr Trp Ser Arg Glu Ser Pro Leu Lys Pro Phe Leu Lys Leu
50 55 60
Ser Leu Gly Leu Pro Gly Leu Gly Ile His Met Arg Pro Leu Ala Ile
65 70 75 80
Trp Leu Phe Ile Phe Asn Val Ser Gln Gln Met Gly Gly Phe Tyr Leu
85 90 95
Cys Gln Pro Gly Pro Pro Ser Glu Lys Ala Trp Gln Pro Gly Trp Thr
100 105 110
Val Asn Val Glu Gly Ser Gly Glu Leu Phe Arg Trp Asn Val Ser Asp
115 120 125
Leu Gly Gly Leu Gly Cys Gly Leu Lys Asn Arg Ser Ser Glu Gly Pro
130 135 140
Ser Ser Pro Ser Gly Lys Leu Met Ser Pro Lys Leu Tyr Val Trp Ala
145 150 155 160
Lys Asp Arg Pro Glu Ile Trp Glu Gly Glu Pro Pro Cys Leu Pro Pro
165 170 175
Arg Asp Ser Leu Asn Gln Ser Leu Ser Gln Asp Leu Thr Met Ala Pro
180 185 190
Gly Ser Thr Leu Trp Leu Ser Cys Gly Val Pro Pro Asp Ser Val Ser
195 200 205
Arg Gly Pro Leu Ser Trp Thr His Val His Pro Lys Gly Pro Lys Ser
210 215 220
Leu Leu Ser Leu Glu Leu Lys Asp Asp Arg Pro Ala Arg Asp Met Trp
225 230 235 240
Val Met Glu Thr Gly Leu Leu Leu Pro Arg Ala Thr Ala Gln Asp Ala
245 250 255
Gly Lys Tyr Tyr Cys His Arg Gly Asn Leu Thr Met Ser Phe His Leu
260 265 270
Glu Ile Thr Ala Arg Pro Val Leu Trp His Trp Leu Leu Arg Thr Gly
275 280 285
Gly Trp Lys Val Ser Ala Val Thr Leu Ala Tyr Leu Ile Phe Cys Leu
290 295 300
Cys Ser Leu Val Gly Ile Leu His Leu Gln Arg Ala Leu Val Leu Arg
305 310 315 320
Arg Lys Arg Lys Arg Met Thr Asp Pro Thr Arg Arg Phe Phe Lys Val
325 330 335
Thr Pro Pro Pro Gly Ser Gly Pro Gln Asn Gln Tyr Gly Asn Val Leu
340 345 350
Ser Leu Pro Thr Pro Thr Ser Gly Leu Gly Arg Ala Gln Arg Trp Ala
355 360 365
Ala Gly Leu Gly Gly Thr Ala Pro Ser Tyr Gly Asn Pro Ser Ser Asp
370 375 380
Val Gln Ala Asp Gly Ala Leu Gly Ser Arg Ser Pro Pro Gly Val Gly
385 390 395 400
Pro Glu Glu Glu Glu Gly Glu Gly Tyr Glu Glu Pro Asp Ser Glu Glu
405 410 415
Asp Ser Glu Phe Tyr Glu Asn Asp Ser Asn Leu Gly Gln Asp Gln Leu
420 425 430
Ser Gln Asp Gly Ser Gly Tyr Glu Asn Pro Glu Asp Glu Pro Leu Gly
435 440 445
Pro Glu Asp Glu Asp Ser Phe Ser Asn Ala Glu Ser Tyr Glu Asn Glu
450 455 460
Asp Glu Glu Leu Thr Gln Pro Val Ala Arg Thr Met Asp Phe Leu Ser
465 470 475 480
Pro His Gly Ser Ala Trp Asp Pro Ser Arg Glu Ala Thr Ser Leu Gly
485 490 495
Ser Gln Ser Tyr Glu Asp Met Arg Gly Ile Leu Tyr Ala Ala Pro Gln
500 505 510
Leu Arg Ser Ile Arg Gly Gln Pro Gly Pro Asn His Glu Glu Asp Ala
515 520 525
Asp Ser Tyr Glu Asn Met Asp Asn Pro Asp Gly Pro Asp Pro Ala Trp
530 535 540
Gly Gly Gly Gly Arg Met Gly Thr Trp Ser Thr Arg
545 550 555
<210> 16
<211> 232
<212> PRT
<213> artificial sequence
<220>
<223> human IgG1 Fc
<400> 16
Glu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala
1 5 10 15
Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro
20 25 30
Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val
35 40 45
Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val
50 55 60
Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln
65 70 75 80
Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln
85 90 95
Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala
100 105 110
Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro
115 120 125
Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr
130 135 140
Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser
145 150 155 160
Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr
165 170 175
Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr
180 185 190
Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe
195 200 205
Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys
210 215 220
Ser Leu Ser Leu Ser Pro Gly Lys
225 230
<210> 17
<211> 231
<212> PRT
<213> artificial sequence
<220>
<223> modified human IgG1 Fc
<400> 17
Pro Lys Ser Ser Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro
1 5 10 15
Glu Ala Glu Gly Ala Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys
20 25 30
Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val
35 40 45
Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp
50 55 60
Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr
65 70 75 80
Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp
85 90 95
Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu
100 105 110
Pro Ser Ser Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg
115 120 125
Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr Lys
130 135 140
Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp
145 150 155 160
Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys
165 170 175
Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser
180 185 190
Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser
195 200 205
Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser
210 215 220
Leu Ser Leu Ser Pro Gly Lys
225 230
<210> 18
<211> 290
<212> PRT
<213> artificial sequence
<220>
<223> BCMA-Fc fusion polypeptide
<400> 18
Met Leu Gln Met Ala Gly Gln Cys Ser Gln Asn Glu Tyr Phe Asp Ser
1 5 10 15
Leu Leu His Ala Cys Ile Pro Cys Gln Leu Arg Cys Ser Ser Asn Thr
20 25 30
Pro Pro Leu Thr Cys Gln Arg Tyr Cys Asn Ala Ser Val Thr Asn Ser
35 40 45
Val Lys Gly Thr Asn Ala Gly Gly Gly Gly Ser Pro Lys Ser Ser Asp
50 55 60
Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Ala Glu Gly Ala
65 70 75 80
Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile
85 90 95
Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu
100 105 110
Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His
115 120 125
Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg
130 135 140
Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys
145 150 155 160
Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ser Ser Ile Glu
165 170 175
Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr
180 185 190
Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu
195 200 205
Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp
210 215 220
Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val
225 230 235 240
Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp
245 250 255
Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His
260 265 270
Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro
275 280 285
Gly Lys
290
<210> 19
<211> 377
<212> PRT
<213> artificial sequence
<220>
<223> human ROR1 fragment
<400> 19
Gln Glu Thr Glu Leu Ser Val Ser Ala Glu Leu Val Pro Thr Ser Ser
1 5 10 15
Trp Asn Ile Ser Ser Glu Leu Asn Lys Asp Ser Tyr Leu Thr Leu Asp
20 25 30
Glu Pro Met Asn Asn Ile Thr Thr Ser Leu Gly Gln Thr Ala Glu Leu
35 40 45
His Cys Lys Val Ser Gly Asn Pro Pro Pro Thr Ile Arg Trp Phe Lys
50 55 60
Asn Asp Ala Pro Val Val Gln Glu Pro Arg Arg Leu Ser Phe Arg Ser
65 70 75 80
Thr Ile Tyr Gly Ser Arg Leu Arg Ile Arg Asn Leu Asp Thr Thr Asp
85 90 95
Thr Gly Tyr Phe Gln Cys Val Ala Thr Asn Gly Lys Glu Val Val Ser
100 105 110
Ser Thr Gly Val Leu Phe Val Lys Phe Gly Pro Pro Pro Thr Ala Ser
115 120 125
Pro Gly Tyr Ser Asp Glu Tyr Glu Glu Asp Gly Phe Cys Gln Pro Tyr
130 135 140
Arg Gly Ile Ala Cys Ala Arg Phe Ile Gly Asn Arg Thr Val Tyr Met
145 150 155 160
Glu Ser Leu His Met Gln Gly Glu Ile Glu Asn Gln Ile Thr Ala Ala
165 170 175
Phe Thr Met Ile Gly Thr Ser Ser His Leu Ser Asp Lys Cys Ser Gln
180 185 190
Phe Ala Ile Pro Ser Leu Cys His Tyr Ala Phe Pro Tyr Cys Asp Glu
195 200 205
Thr Ser Ser Val Pro Lys Pro Arg Asp Leu Cys Arg Asp Glu Cys Glu
210 215 220
Ile Leu Glu Asn Val Leu Cys Gln Thr Glu Tyr Ile Phe Ala Arg Ser
225 230 235 240
Asn Pro Met Ile Leu Met Arg Leu Lys Leu Pro Asn Cys Glu Asp Leu
245 250 255
Pro Gln Pro Glu Ser Pro Glu Ala Ala Asn Cys Ile Arg Ile Gly Ile
260 265 270
Pro Met Ala Asp Pro Ile Asn Lys Asn His Lys Cys Tyr Asn Ser Thr
275 280 285
Gly Val Asp Tyr Arg Gly Thr Val Ser Val Thr Lys Ser Gly Arg Gln
290 295 300
Cys Gln Pro Trp Asn Ser Gln Tyr Pro His Thr His Thr Phe Thr Ala
305 310 315 320
Leu Arg Phe Pro Glu Leu Asn Gly Gly His Ser Tyr Cys Arg Asn Pro
325 330 335
Gly Asn Gln Lys Glu Ala Pro Trp Cys Phe Thr Leu Asp Glu Asn Phe
340 345 350
Lys Ser Asp Leu Cys Asp Ile Pro Ala Cys Asp Ser Lys Asp Ser Lys
355 360 365
Glu Lys Asn Lys Met Glu Ile Leu Tyr
370 375
<210> 20
<211> 613
<212> PRT
<213> artificial sequence
<220>
<223> ROR1-Fc fusion polypeptide
<400> 20
Gln Glu Thr Glu Leu Ser Val Ser Ala Glu Leu Val Pro Thr Ser Ser
1 5 10 15
Trp Asn Ile Ser Ser Glu Leu Asn Lys Asp Ser Tyr Leu Thr Leu Asp
20 25 30
Glu Pro Met Asn Asn Ile Thr Thr Ser Leu Gly Gln Thr Ala Glu Leu
35 40 45
His Cys Lys Val Ser Gly Asn Pro Pro Pro Thr Ile Arg Trp Phe Lys
50 55 60
Asn Asp Ala Pro Val Val Gln Glu Pro Arg Arg Leu Ser Phe Arg Ser
65 70 75 80
Thr Ile Tyr Gly Ser Arg Leu Arg Ile Arg Asn Leu Asp Thr Thr Asp
85 90 95
Thr Gly Tyr Phe Gln Cys Val Ala Thr Asn Gly Lys Glu Val Val Ser
100 105 110
Ser Thr Gly Val Leu Phe Val Lys Phe Gly Pro Pro Pro Thr Ala Ser
115 120 125
Pro Gly Tyr Ser Asp Glu Tyr Glu Glu Asp Gly Phe Cys Gln Pro Tyr
130 135 140
Arg Gly Ile Ala Cys Ala Arg Phe Ile Gly Asn Arg Thr Val Tyr Met
145 150 155 160
Glu Ser Leu His Met Gln Gly Glu Ile Glu Asn Gln Ile Thr Ala Ala
165 170 175
Phe Thr Met Ile Gly Thr Ser Ser His Leu Ser Asp Lys Cys Ser Gln
180 185 190
Phe Ala Ile Pro Ser Leu Cys His Tyr Ala Phe Pro Tyr Cys Asp Glu
195 200 205
Thr Ser Ser Val Pro Lys Pro Arg Asp Leu Cys Arg Asp Glu Cys Glu
210 215 220
Ile Leu Glu Asn Val Leu Cys Gln Thr Glu Tyr Ile Phe Ala Arg Ser
225 230 235 240
Asn Pro Met Ile Leu Met Arg Leu Lys Leu Pro Asn Cys Glu Asp Leu
245 250 255
Pro Gln Pro Glu Ser Pro Glu Ala Ala Asn Cys Ile Arg Ile Gly Ile
260 265 270
Pro Met Ala Asp Pro Ile Asn Lys Asn His Lys Cys Tyr Asn Ser Thr
275 280 285
Gly Val Asp Tyr Arg Gly Thr Val Ser Val Thr Lys Ser Gly Arg Gln
290 295 300
Cys Gln Pro Trp Asn Ser Gln Tyr Pro His Thr His Thr Phe Thr Ala
305 310 315 320
Leu Arg Phe Pro Glu Leu Asn Gly Gly His Ser Tyr Cys Arg Asn Pro
325 330 335
Gly Asn Gln Lys Glu Ala Pro Trp Cys Phe Thr Leu Asp Glu Asn Phe
340 345 350
Lys Ser Asp Leu Cys Asp Ile Pro Ala Cys Asp Ser Lys Asp Ser Lys
355 360 365
Glu Lys Asn Lys Met Glu Ile Leu Tyr Gly Gly Gly Gly Ser Pro Lys
370 375 380
Ser Ser Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Ala
385 390 395 400
Glu Gly Ala Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr
405 410 415
Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val
420 425 430
Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val
435 440 445
Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser
450 455 460
Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu
465 470 475 480
Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ser
485 490 495
Ser Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro
500 505 510
Gln Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln
515 520 525
Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala
530 535 540
Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr
545 550 555 560
Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu
565 570 575
Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser
580 585 590
Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser
595 600 605
Leu Ser Pro Gly Lys
610
<210> 21
<211> 904
<212> PRT
<213> artificial sequence
<220>
<223> CD22-Fc fusion polypeptide
<400> 21
Asp Ser Ser Lys Trp Val Phe Glu His Pro Glu Thr Leu Tyr Ala Trp
1 5 10 15
Glu Gly Ala Cys Val Trp Ile Pro Cys Thr Tyr Arg Ala Leu Asp Gly
20 25 30
Asp Leu Glu Ser Phe Ile Leu Phe His Asn Pro Glu Tyr Asn Lys Asn
35 40 45
Thr Ser Lys Phe Asp Gly Thr Arg Leu Tyr Glu Ser Thr Lys Asp Gly
50 55 60
Lys Val Pro Ser Glu Gln Lys Arg Val Gln Phe Leu Gly Asp Lys Asn
65 70 75 80
Lys Asn Cys Thr Leu Ser Ile His Pro Val His Leu Asn Asp Ser Gly
85 90 95
Gln Leu Gly Leu Arg Met Glu Ser Lys Thr Glu Lys Trp Met Glu Arg
100 105 110
Ile His Leu Asn Val Ser Glu Arg Pro Phe Pro Pro His Ile Gln Leu
115 120 125
Pro Pro Glu Ile Gln Glu Ser Gln Glu Val Thr Leu Thr Cys Leu Leu
130 135 140
Asn Phe Ser Cys Tyr Gly Tyr Pro Ile Gln Leu Gln Trp Leu Leu Glu
145 150 155 160
Gly Val Pro Met Arg Gln Ala Ala Val Thr Ser Thr Ser Leu Thr Ile
165 170 175
Lys Ser Val Phe Thr Arg Ser Glu Leu Lys Phe Ser Pro Gln Trp Ser
180 185 190
His His Gly Lys Ile Val Thr Cys Gln Leu Gln Asp Ala Asp Gly Lys
195 200 205
Phe Leu Ser Asn Asp Thr Val Gln Leu Asn Val Lys His Thr Pro Lys
210 215 220
Leu Glu Ile Lys Val Thr Pro Ser Asp Ala Ile Val Arg Glu Gly Asp
225 230 235 240
Ser Val Thr Met Thr Cys Glu Val Ser Ser Ser Asn Pro Glu Tyr Thr
245 250 255
Thr Val Ser Trp Leu Lys Asp Gly Thr Ser Leu Lys Lys Gln Asn Thr
260 265 270
Phe Thr Leu Asn Leu Arg Glu Val Thr Lys Asp Gln Ser Gly Lys Tyr
275 280 285
Cys Cys Gln Val Ser Asn Asp Val Gly Pro Gly Arg Ser Glu Glu Val
290 295 300
Phe Leu Gln Val Gln Tyr Ala Pro Glu Pro Ser Thr Val Gln Ile Leu
305 310 315 320
His Ser Pro Ala Val Glu Gly Ser Gln Val Glu Phe Leu Cys Met Ser
325 330 335
Leu Ala Asn Pro Leu Pro Thr Asn Tyr Thr Trp Tyr His Asn Gly Lys
340 345 350
Glu Met Gln Gly Arg Thr Glu Glu Lys Val His Ile Pro Lys Ile Leu
355 360 365
Pro Trp His Ala Gly Thr Tyr Ser Cys Val Ala Glu Asn Ile Leu Gly
370 375 380
Thr Gly Gln Arg Gly Pro Gly Ala Glu Leu Asp Val Gln Tyr Pro Pro
385 390 395 400
Lys Lys Val Thr Thr Val Ile Gln Asn Pro Met Pro Ile Arg Glu Gly
405 410 415
Asp Thr Val Thr Leu Ser Cys Asn Tyr Asn Ser Ser Asn Pro Ser Val
420 425 430
Thr Arg Tyr Glu Trp Lys Pro His Gly Ala Trp Glu Glu Pro Ser Leu
435 440 445
Gly Val Leu Lys Ile Gln Asn Val Gly Trp Asp Asn Thr Thr Ile Ala
450 455 460
Cys Ala Ala Cys Asn Ser Trp Cys Ser Trp Ala Ser Pro Val Ala Leu
465 470 475 480
Asn Val Gln Tyr Ala Pro Arg Asp Val Arg Val Arg Lys Ile Lys Pro
485 490 495
Leu Ser Glu Ile His Ser Gly Asn Ser Val Ser Leu Gln Cys Asp Phe
500 505 510
Ser Ser Ser His Pro Lys Glu Val Gln Phe Phe Trp Glu Lys Asn Gly
515 520 525
Arg Leu Leu Gly Lys Glu Ser Gln Leu Asn Phe Asp Ser Ile Ser Pro
530 535 540
Glu Asp Ala Gly Ser Tyr Ser Cys Trp Val Asn Asn Ser Ile Gly Gln
545 550 555 560
Thr Ala Ser Lys Ala Trp Thr Leu Glu Val Leu Tyr Ala Pro Arg Arg
565 570 575
Leu Arg Val Ser Met Ser Pro Gly Asp Gln Val Met Glu Gly Lys Ser
580 585 590
Ala Thr Leu Thr Cys Glu Ser Asp Ala Asn Pro Pro Val Ser His Tyr
595 600 605
Thr Trp Phe Asp Trp Asn Asn Gln Ser Leu Pro Tyr His Ser Gln Lys
610 615 620
Leu Arg Leu Glu Pro Val Lys Val Gln His Ser Gly Ala Tyr Trp Cys
625 630 635 640
Gln Gly Thr Asn Ser Val Gly Lys Gly Arg Ser Pro Leu Ser Thr Leu
645 650 655
Thr Val Tyr Tyr Ser Pro Glu Thr Ile Gly Arg Arg Gly Gly Gly Gly
660 665 670
Ser Pro Lys Ser Ser Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala
675 680 685
Pro Glu Ala Glu Gly Ala Pro Ser Val Phe Leu Phe Pro Pro Lys Pro
690 695 700
Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val
705 710 715 720
Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val
725 730 735
Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln
740 745 750
Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln
755 760 765
Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala
770 775 780
Leu Pro Ser Ser Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro
785 790 795 800
Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr
805 810 815
Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser
820 825 830
Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr
835 840 845
Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr
850 855 860
Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe
865 870 875 880
Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys
885 890 895
Ser Leu Ser Leu Ser Pro Gly Lys
900
<210> 22
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> joint
<400> 22
Gly Gly Gly Gly Ser
1 5
<210> 23
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> joint
<400> 23
Gly Gly Gly Ser
1
<210> 24
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> joint
<400> 24
Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser
1 5 10 15
<210> 25
<211> 18
<212> PRT
<213> artificial sequence
<220>
<223> joint
<400> 25
Gly Ser Thr Ser Gly Ser Gly Lys Pro Gly Ser Gly Glu Gly Ser Thr
1 5 10 15
Lys Gly
<210> 26
<211> 21
<212> PRT
<213> artificial sequence
<220>
<223> joint
<400> 26
Ser Arg Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly
1 5 10 15
Ser Leu Glu Met Ala
20
<210> 27
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> CDR H1
<400> 27
Asp Tyr Gly Val Ser
1 5
<210> 28
<211> 16
<212> PRT
<213> artificial sequence
<220>
<223> CDR H2
<400> 28
Val Ile Trp Gly Ser Glu Thr Thr Tyr Tyr Asn Ser Ala Leu Lys Ser
1 5 10 15
<210> 29
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> CDR H3
<400> 29
Tyr Ala Met Asp Tyr Trp Gly
1 5
<210> 30
<211> 12
<212> PRT
<213> artificial sequence
<220>
<223> CDR H3
<400> 30
His Tyr Tyr Tyr Gly Gly Ser Tyr Ala Met Asp Tyr
1 5 10
<210> 31
<211> 11
<212> PRT
<213> artificial sequence
<220>
<223> CDR L1
<400> 31
Arg Ala Ser Gln Asp Ile Ser Lys Tyr Leu Asn
1 5 10
<210> 32
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> CDR L2
<400> 32
Ser Arg Leu His Ser Gly Val
1 5
<210> 33
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> CDR L2
<400> 33
His Thr Ser Arg Leu His Ser
1 5
<210> 34
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> CDR L3
<400> 34
Gly Asn Thr Leu Pro Tyr Thr Phe Gly
1 5
<210> 35
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> CDR L3
<400> 35
Gln Gln Gly Asn Thr Leu Pro Tyr Thr
1 5
<210> 36
<211> 120
<212> PRT
<213> artificial sequence
<220>
<223> VH
<400> 36
Glu Val Lys Leu Gln Glu Ser Gly Pro Gly Leu Val Ala Pro Ser Gln
1 5 10 15
Ser Leu Ser Val Thr Cys Thr Val Ser Gly Val Ser Leu Pro Asp Tyr
20 25 30
Gly Val Ser Trp Ile Arg Gln Pro Pro Arg Lys Gly Leu Glu Trp Leu
35 40 45
Gly Val Ile Trp Gly Ser Glu Thr Thr Tyr Tyr Asn Ser Ala Leu Lys
50 55 60
Ser Arg Leu Thr Ile Ile Lys Asp Asn Ser Lys Ser Gln Val Phe Leu
65 70 75 80
Lys Met Asn Ser Leu Gln Thr Asp Asp Thr Ala Ile Tyr Tyr Cys Ala
85 90 95
Lys His Tyr Tyr Tyr Gly Gly Ser Tyr Ala Met Asp Tyr Trp Gly Gln
100 105 110
Gly Thr Ser Val Thr Val Ser Ser
115 120
<210> 37
<211> 107
<212> PRT
<213> artificial sequence
<220>
<223> VL
<400> 37
Asp Ile Gln Met Thr Gln Thr Thr Ser Ser Leu Ser Ala Ser Leu Gly
1 5 10 15
Asp Arg Val Thr Ile Ser Cys Arg Ala Ser Gln Asp Ile Ser Lys Tyr
20 25 30
Leu Asn Trp Tyr Gln Gln Lys Pro Asp Gly Thr Val Lys Leu Leu Ile
35 40 45
Tyr His Thr Ser Arg Leu His Ser Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Tyr Ser Leu Thr Ile Ser Asn Leu Glu Gln
65 70 75 80
Glu Asp Ile Ala Thr Tyr Phe Cys Gln Gln Gly Asn Thr Leu Pro Tyr
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Thr
100 105
<210> 38
<211> 735
<212> DNA
<213> artificial sequence
<220>
<223> sequence encoding scFv
<400> 38
gacatccaga tgacccagac cacctccagc ctgagcgcca gcctgggcga ccgggtgacc 60
atcagctgcc gggccagcca ggacatcagc aagtacctga actggtatca gcagaagccc 120
gacggcaccg tcaagctgct gatctaccac accagccggc tgcacagcgg cgtgcccagc 180
cggtttagcg gcagcggctc cggcaccgac tacagcctga ccatctccaa cctggaacag 240
gaagatatcg ccacctactt ttgccagcag ggcaacacac tgccctacac ctttggcggc 300
ggaacaaagc tggaaatcac cggcagcacc tccggcagcg gcaagcctgg cagcggcgag 360
ggcagcacca agggcgaggt gaagctgcag gaaagcggcc ctggcctggt ggcccccagc 420
cagagcctga gcgtgacctg caccgtgagc ggcgtgagcc tgcccgacta cggcgtgagc 480
tggatccggc agccccccag gaagggcctg gaatggctgg gcgtgatctg gggcagcgag 540
accacctact acaacagcgc cctgaagagc cggctgacca tcatcaagga caacagcaag 600
agccaggtgt tcctgaagat gaacagcctg cagaccgacg acaccgccat ctactactgc 660
gccaagcact actactacgg cggcagctac gccatggact actggggcca gggcaccagc 720
gtgaccgtga gcagc 735
<210> 39
<211> 245
<212> PRT
<213> artificial sequence
<220>
<223> scFv
<400> 39
Asp Ile Gln Met Thr Gln Thr Thr Ser Ser Leu Ser Ala Ser Leu Gly
1 5 10 15
Asp Arg Val Thr Ile Ser Cys Arg Ala Ser Gln Asp Ile Ser Lys Tyr
20 25 30
Leu Asn Trp Tyr Gln Gln Lys Pro Asp Gly Thr Val Lys Leu Leu Ile
35 40 45
Tyr His Thr Ser Arg Leu His Ser Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Tyr Ser Leu Thr Ile Ser Asn Leu Glu Gln
65 70 75 80
Glu Asp Ile Ala Thr Tyr Phe Cys Gln Gln Gly Asn Thr Leu Pro Tyr
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Thr Gly Ser Thr Ser Gly
100 105 110
Ser Gly Lys Pro Gly Ser Gly Glu Gly Ser Thr Lys Gly Glu Val Lys
115 120 125
Leu Gln Glu Ser Gly Pro Gly Leu Val Ala Pro Ser Gln Ser Leu Ser
130 135 140
Val Thr Cys Thr Val Ser Gly Val Ser Leu Pro Asp Tyr Gly Val Ser
145 150 155 160
Trp Ile Arg Gln Pro Pro Arg Lys Gly Leu Glu Trp Leu Gly Val Ile
165 170 175
Trp Gly Ser Glu Thr Thr Tyr Tyr Asn Ser Ala Leu Lys Ser Arg Leu
180 185 190
Thr Ile Ile Lys Asp Asn Ser Lys Ser Gln Val Phe Leu Lys Met Asn
195 200 205
Ser Leu Gln Thr Asp Asp Thr Ala Ile Tyr Tyr Cys Ala Lys His Tyr
210 215 220
Tyr Tyr Gly Gly Ser Tyr Ala Met Asp Tyr Trp Gly Gln Gly Thr Ser
225 230 235 240
Val Thr Val Ser Ser
245
<210> 40
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> CDR H1
<400> 40
Ser Tyr Trp Met Asn
1 5
<210> 41
<211> 17
<212> PRT
<213> artificial sequence
<220>
<223> CDR H2
<400> 41
Gln Ile Tyr Pro Gly Asp Gly Asp Thr Asn Tyr Asn Gly Lys Phe Lys
1 5 10 15
Gly
<210> 42
<211> 13
<212> PRT
<213> artificial sequence
<220>
<223> CDR H3
<400> 42
Lys Thr Ile Ser Ser Val Val Asp Phe Tyr Phe Asp Tyr
1 5 10
<210> 43
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> CDR L1
<400> 43
Gln Gln Tyr Asn Arg Tyr Pro Tyr Thr
1 5
<210> 44
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> CDR L2
<400> 44
Ser Tyr Trp Met Asn
1 5
<210> 45
<211> 17
<212> PRT
<213> artificial sequence
<220>
<223> CDR L3
<400> 45
Gln Ile Tyr Pro Gly Asp Gly Asp Thr Asn Tyr Asn Gly Lys Phe Lys
1 5 10 15
Gly
<210> 46
<211> 122
<212> PRT
<213> artificial sequence
<220>
<223> VH
<400> 46
Glu Val Lys Leu Gln Gln Ser Gly Ala Glu Leu Val Arg Pro Gly Ser
1 5 10 15
Ser Val Lys Ile Ser Cys Lys Ala Ser Gly Tyr Ala Phe Ser Ser Tyr
20 25 30
Trp Met Asn Trp Val Lys Gln Arg Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Gln Ile Tyr Pro Gly Asp Gly Asp Thr Asn Tyr Asn Gly Lys Phe
50 55 60
Lys Gly Gln Ala Thr Leu Thr Ala Asp Lys Ser Ser Ser Thr Ala Tyr
65 70 75 80
Met Gln Leu Ser Gly Leu Thr Ser Glu Asp Ser Ala Val Tyr Phe Cys
85 90 95
Ala Arg Lys Thr Ile Ser Ser Val Val Asp Phe Tyr Phe Asp Tyr Trp
100 105 110
Gly Gln Gly Thr Thr Val Thr Val Ser Ser
115 120
<210> 47
<211> 108
<212> PRT
<213> artificial sequence
<220>
<223> VL
<400> 47
Asp Ile Glu Leu Thr Gln Ser Pro Lys Phe Met Ser Thr Ser Val Gly
1 5 10 15
Asp Arg Val Ser Val Thr Cys Lys Ala Ser Gln Asn Val Gly Thr Asn
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ser Pro Lys Pro Leu Ile
35 40 45
Tyr Ser Ala Thr Tyr Arg Asn Ser Gly Val Pro Asp Arg Phe Thr Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Thr Asn Val Gln Ser
65 70 75 80
Lys Asp Leu Ala Asp Tyr Phe Cys Gln Gln Tyr Asn Arg Tyr Pro Tyr
85 90 95
Thr Ser Gly Gly Gly Thr Lys Leu Glu Ile Lys Arg
100 105
<210> 48
<211> 11
<212> PRT
<213> artificial sequence
<220>
<223> CDR L1
<400> 48
Lys Ala Ser Gln Asn Val Gly Thr Asn Val Ala
1 5 10
<210> 49
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> CDR L2
<400> 49
Ser Ala Thr Tyr Arg Asn Ser
1 5
<210> 50
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> CDR L3
<400> 50
Gln Gln Tyr Asn Arg Tyr Pro Tyr Thr
1 5
<210> 51
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> CDR H1
<400> 51
Ser Tyr Trp Met Asn
1 5
<210> 52
<211> 17
<212> PRT
<213> artificial sequence
<220>
<223> CDR H2
<400> 52
Gln Ile Tyr Pro Gly Asp Gly Asp Thr Asn Tyr Asn Gly Lys Phe Lys
1 5 10 15
Gly
<210> 53
<211> 13
<212> PRT
<213> artificial sequence
<220>
<223> CDR H3
<400> 53
Lys Thr Ile Ser Ser Val Val Asp Phe Tyr Phe Asp Tyr
1 5 10
<210> 54
<211> 245
<212> PRT
<213> artificial sequence
<220>
<223> scFv
<400> 54
Glu Val Lys Leu Gln Gln Ser Gly Ala Glu Leu Val Arg Pro Gly Ser
1 5 10 15
Ser Val Lys Ile Ser Cys Lys Ala Ser Gly Tyr Ala Phe Ser Ser Tyr
20 25 30
Trp Met Asn Trp Val Lys Gln Arg Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Gln Ile Tyr Pro Gly Asp Gly Asp Thr Asn Tyr Asn Gly Lys Phe
50 55 60
Lys Gly Gln Ala Thr Leu Thr Ala Asp Lys Ser Ser Ser Thr Ala Tyr
65 70 75 80
Met Gln Leu Ser Gly Leu Thr Ser Glu Asp Ser Ala Val Tyr Phe Cys
85 90 95
Ala Arg Lys Thr Ile Ser Ser Val Val Asp Phe Tyr Phe Asp Tyr Trp
100 105 110
Gly Gln Gly Thr Thr Val Thr Val Ser Ser Gly Gly Gly Gly Ser Gly
115 120 125
Gly Gly Gly Ser Gly Gly Gly Gly Ser Asp Ile Glu Leu Thr Gln Ser
130 135 140
Pro Lys Phe Met Ser Thr Ser Val Gly Asp Arg Val Ser Val Thr Cys
145 150 155 160
Lys Ala Ser Gln Asn Val Gly Thr Asn Val Ala Trp Tyr Gln Gln Lys
165 170 175
Pro Gly Gln Ser Pro Lys Pro Leu Ile Tyr Ser Ala Thr Tyr Arg Asn
180 185 190
Ser Gly Val Pro Asp Arg Phe Thr Gly Ser Gly Ser Gly Thr Asp Phe
195 200 205
Thr Leu Thr Ile Thr Asn Val Gln Ser Lys Asp Leu Ala Asp Tyr Phe
210 215 220
Cys Gln Gln Tyr Asn Arg Tyr Pro Tyr Thr Ser Gly Gly Gly Thr Lys
225 230 235 240
Leu Glu Ile Lys Arg
245
<210> 55
<211> 117
<212> PRT
<213> artificial sequence
<220>
<223> VH
<400> 55
Gln Ile Gln Leu Val Gln Ser Gly Pro Glu Leu Lys Lys Pro Gly Glu
1 5 10 15
Thr Val Lys Ile Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Asp Tyr
20 25 30
Ser Ile Asn Trp Val Lys Arg Ala Pro Gly Lys Gly Leu Lys Trp Met
35 40 45
Gly Trp Ile Asn Thr Glu Thr Arg Glu Pro Ala Tyr Ala Tyr Asp Phe
50 55 60
Arg Gly Arg Phe Ala Phe Ser Leu Glu Thr Ser Ala Ser Thr Ala Tyr
65 70 75 80
Leu Gln Ile Asn Asn Leu Lys Tyr Glu Asp Thr Ala Thr Tyr Phe Cys
85 90 95
Ala Leu Asp Tyr Ser Tyr Ala Met Asp Tyr Trp Gly Gln Gly Thr Ser
100 105 110
Val Thr Val Ser Ser
115
<210> 56
<211> 111
<212> PRT
<213> artificial sequence
<220>
<223> VL
<400> 56
Asp Ile Val Leu Thr Gln Ser Pro Pro Ser Leu Ala Met Ser Leu Gly
1 5 10 15
Lys Arg Ala Thr Ile Ser Cys Arg Ala Ser Glu Ser Val Thr Ile Leu
20 25 30
Gly Ser His Leu Ile His Trp Tyr Gln Gln Lys Pro Gly Gln Pro Pro
35 40 45
Thr Leu Leu Ile Gln Leu Ala Ser Asn Val Gln Thr Gly Val Pro Ala
50 55 60
Arg Phe Ser Gly Ser Gly Ser Arg Thr Asp Phe Thr Leu Thr Ile Asp
65 70 75 80
Pro Val Glu Glu Asp Asp Val Ala Val Tyr Tyr Cys Leu Gln Ser Arg
85 90 95
Thr Ile Pro Arg Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105 110
<210> 57
<211> 122
<212> PRT
<213> artificial sequence
<220>
<223> VH
<400> 57
Gln Ile Gln Leu Val Gln Ser Gly Pro Asp Leu Lys Lys Pro Gly Glu
1 5 10 15
Thr Val Lys Leu Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Asn Phe
20 25 30
Gly Met Asn Trp Val Lys Gln Ala Pro Gly Lys Gly Phe Lys Trp Met
35 40 45
Ala Trp Ile Asn Thr Tyr Thr Gly Glu Ser Tyr Phe Ala Asp Asp Phe
50 55 60
Lys Gly Arg Phe Ala Phe Ser Val Glu Thr Ser Ala Thr Thr Ala Tyr
65 70 75 80
Leu Gln Ile Asn Asn Leu Lys Thr Glu Asp Thr Ala Thr Tyr Phe Cys
85 90 95
Ala Arg Gly Glu Ile Tyr Tyr Gly Tyr Asp Gly Gly Phe Ala Tyr Trp
100 105 110
Gly Gln Gly Thr Leu Val Thr Val Ser Ala
115 120
<210> 58
<211> 107
<212> PRT
<213> artificial sequence
<220>
<223> VL
<400> 58
Asp Val Val Met Thr Gln Ser His Arg Phe Met Ser Thr Ser Val Gly
1 5 10 15
Asp Arg Val Ser Ile Thr Cys Arg Ala Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ser Trp Tyr Gln Gln Lys Pro Gly Gln Ser Pro Lys Leu Leu Ile
35 40 45
Phe Ser Ala Ser Tyr Arg Tyr Thr Gly Val Pro Asp Arg Phe Thr Gly
50 55 60
Ser Gly Ser Gly Ala Asp Phe Thr Leu Thr Ile Ser Ser Val Gln Ala
65 70 75 80
Glu Asp Leu Ala Val Tyr Tyr Cys Gln Gln His Tyr Ser Thr Pro Trp
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Asp Ile Lys
100 105
<210> 59
<211> 116
<212> PRT
<213> artificial sequence
<220>
<223> VH
<400> 59
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Trp Ile Gly Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Ile Ile Tyr Pro Gly Asp Ser Asp Thr Arg Tyr Ser Pro Ser Phe
50 55 60
Gln Gly His Val Thr Ile Ser Ala Asp Lys Ser Ile Ser Thr Ala Tyr
65 70 75 80
Leu Gln Trp Ser Ser Leu Lys Ala Ser Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Ala Arg Tyr Ser Gly Ser Phe Asp Asn Trp Gly Gln Gly Thr Leu Val
100 105 110
Thr Val Ser Ser
115
<210> 60
<211> 111
<212> PRT
<213> artificial sequence
<220>
<223> VL
<400> 60
Ser Tyr Glu Leu Thr Gln Pro Pro Ser Ala Ser Gly Thr Pro Gly Gln
1 5 10 15
Arg Val Thr Met Ser Cys Ser Gly Thr Ser Ser Asn Ile Gly Ser His
20 25 30
Ser Val Asn Trp Tyr Gln Gln Leu Pro Gly Thr Ala Pro Lys Leu Leu
35 40 45
Ile Tyr Thr Asn Asn Gln Arg Pro Ser Gly Val Pro Asp Arg Phe Ser
50 55 60
Gly Ser Lys Ser Gly Thr Ser Ala Ser Leu Ala Ile Ser Gly Leu Gln
65 70 75 80
Ser Glu Asp Glu Ala Asp Tyr Tyr Cys Ala Ala Trp Asp Gly Ser Leu
85 90 95
Asn Gly Leu Val Phe Gly Gly Gly Thr Lys Leu Thr Val Leu Gly
100 105 110
<210> 61
<211> 118
<212> PRT
<213> artificial sequence
<220>
<223> VH
<400> 61
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Met Lys Lys Pro Gly Ala
1 5 10 15
Ser Leu Lys Leu Ser Cys Lys Ala Ser Gly Tyr Thr Phe Ile Asp Tyr
20 25 30
Tyr Val Tyr Trp Met Arg Gln Ala Pro Gly Gln Gly Leu Glu Ser Met
35 40 45
Gly Trp Ile Asn Pro Asn Ser Gly Gly Thr Asn Tyr Ala Gln Lys Phe
50 55 60
Gln Gly Arg Val Thr Met Thr Arg Asp Thr Ser Ile Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Arg Leu Arg Ser Asp Asp Thr Ala Met Tyr Tyr Cys
85 90 95
Ala Arg Ser Gln Arg Asp Gly Tyr Met Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 62
<211> 105
<212> PRT
<213> artificial sequence
<220>
<223> VL
<400> 62
Gln Ser Ala Leu Thr Gln Pro Ala Ser Val Ser Ala Ser Pro Gly Gln
1 5 10 15
Ser Ile Ala Ile Ser Cys Thr Gly Thr Ser Ser Asp Val Gly Trp Tyr
20 25 30
Gln Gln His Pro Gly Lys Ala Pro Lys Leu Met Ile Tyr Glu Asp Ser
35 40 45
Lys Arg Pro Ser Gly Val Ser Asn Arg Phe Ser Gly Ser Lys Ser Gly
50 55 60
Asn Thr Ala Ser Leu Thr Ile Ser Gly Leu Gln Ala Glu Asp Glu Ala
65 70 75 80
Asp Tyr Tyr Cys Ser Ser Asn Thr Arg Ser Ser Thr Leu Val Phe Gly
85 90 95
Gly Gly Thr Lys Leu Thr Val Leu Gly
100 105
<210> 63
<211> 122
<212> PRT
<213> artificial sequence
<220>
<223> VH
<400> 63
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ser
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Ser Ser Tyr
20 25 30
Ala Ile Ser Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Met
35 40 45
Gly Arg Ile Ile Pro Ile Leu Gly Ile Ala Asn Tyr Ala Gln Lys Phe
50 55 60
Gln Gly Arg Val Thr Met Thr Glu Asp Thr Ser Thr Asp Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Ser Gly Tyr Ser Lys Ser Ile Val Ser Tyr Met Asp Tyr Trp
100 105 110
Gly Gln Gly Thr Leu Val Thr Val Ser Ser
115 120
<210> 64
<211> 111
<212> PRT
<213> artificial sequence
<220>
<223> VL
<400> 64
Leu Pro Val Leu Thr Gln Pro Pro Ser Thr Ser Gly Thr Pro Gly Gln
1 5 10 15
Arg Val Thr Val Ser Cys Ser Gly Ser Ser Ser Asn Ile Gly Ser Asn
20 25 30
Val Val Phe Trp Tyr Gln Gln Leu Pro Gly Thr Ala Pro Lys Leu Val
35 40 45
Ile Tyr Arg Asn Asn Gln Arg Pro Ser Gly Val Pro Asp Arg Phe Ser
50 55 60
Val Ser Lys Ser Gly Thr Ser Ala Ser Leu Ala Ile Ser Gly Leu Arg
65 70 75 80
Ser Glu Asp Glu Ala Asp Tyr Tyr Cys Ala Ala Trp Asp Asp Ser Leu
85 90 95
Ser Gly Tyr Val Phe Gly Thr Gly Thr Lys Val Thr Val Leu Gly
100 105 110
<210> 65
<211> 120
<212> PRT
<213> artificial sequence
<220>
<223> VH
<400> 65
Gln Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ser
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Ser Ser Tyr
20 25 30
Ala Ile Ser Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Met
35 40 45
Gly Arg Ile Ile Pro Ile Leu Gly Thr Ala Asn Tyr Ala Gln Lys Phe
50 55 60
Gln Gly Arg Val Thr Ile Thr Ala Asp Glu Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Ser Gly Tyr Gly Ser Tyr Arg Trp Glu Asp Ser Trp Gly Gln
100 105 110
Gly Thr Leu Val Thr Val Ser Ser
115 120
<210> 66
<211> 112
<212> PRT
<213> artificial sequence
<220>
<223> VL
<400> 66
Gln Ala Val Leu Thr Gln Pro Pro Ser Ala Ser Gly Thr Pro Gly Gln
1 5 10 15
Arg Val Thr Ile Ser Cys Ser Gly Ser Ser Ser Asn Ile Gly Ser Asn
20 25 30
Tyr Val Phe Trp Tyr Gln Gln Leu Pro Gly Thr Ala Pro Lys Leu Leu
35 40 45
Ile Tyr Ser Asn Asn Gln Arg Pro Ser Gly Val Pro Asp Arg Phe Ser
50 55 60
Gly Ser Lys Ser Gly Thr Ser Ala Ser Leu Ala Ile Ser Gly Leu Arg
65 70 75 80
Ser Glu Asp Glu Ala Asp Tyr Tyr Cys Ala Ala Trp Asp Asp Ser Leu
85 90 95
Ser Ala Ser Tyr Val Phe Gly Thr Gly Thr Lys Val Thr Val Leu Gly
100 105 110
<210> 67
<211> 118
<212> PRT
<213> artificial sequence
<220>
<223> VH
<400> 67
Gln Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Asp Tyr
20 25 30
Tyr Met His Trp Val Arg Gln Ala Pro Gly Gln Arg Leu Glu Trp Met
35 40 45
Gly Trp Ile Asn Pro Asn Ser Gly Gly Thr Asn Tyr Ala Gln Lys Phe
50 55 60
Gln Asp Arg Ile Thr Val Thr Arg Asp Thr Ser Ser Asn Thr Gly Tyr
65 70 75 80
Met Glu Leu Thr Arg Leu Arg Ser Asp Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Ser Pro Tyr Ser Gly Val Leu Asp Lys Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 68
<211> 112
<212> PRT
<213> artificial sequence
<220>
<223> VL
<400> 68
Gln Ser Val Leu Thr Gln Pro Pro Ser Val Ser Gly Ala Pro Gly Gln
1 5 10 15
Arg Val Thr Ile Ser Cys Thr Gly Ser Ser Ser Asn Ile Gly Ala Gly
20 25 30
Phe Asp Val His Trp Tyr Gln Gln Leu Pro Gly Thr Ala Pro Lys Leu
35 40 45
Leu Ile Tyr Gly Asn Ser Asn Arg Pro Ser Gly Val Pro Asp Arg Phe
50 55 60
Ser Gly Ser Lys Ser Gly Thr Ser Ala Ser Leu Ala Ile Thr Gly Leu
65 70 75 80
Gln Ala Glu Asp Glu Ala Asp Tyr Tyr Cys Gln Ser Tyr Asp Ser Ser
85 90 95
Leu Ser Gly Tyr Val Phe Gly Thr Gly Thr Lys Val Thr Val Leu Gly
100 105 110
<210> 69
<211> 12
<212> PRT
<213> artificial sequence
<220>
<223> spacer
(IgG 4 hinge) (aa)
<400> 69
Glu Ser Lys Tyr Gly Pro Pro Cys Pro Pro Cys Pro
1 5 10
<210> 70
<211> 36
<212> DNA
<213> artificial sequence
<220>
<223> spacer
(IgG 4 hinge) (nt)
<400> 70
gaatctaagt acggaccgcc ctgcccccct tgccct 36
<210> 71
<211> 119
<212> PRT
<213> Homo Sapiens (Homo Sapiens)
<220>
<223> hinge-CH 3
Spacer
<400> 71
Glu Ser Lys Tyr Gly Pro Pro Cys Pro Pro Cys Pro Gly Gln Pro Arg
1 5 10 15
Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Gln Glu Glu Met Thr Lys
20 25 30
Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp
35 40 45
Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys
50 55 60
Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser
65 70 75 80
Arg Leu Thr Val Asp Lys Ser Arg Trp Gln Glu Gly Asn Val Phe Ser
85 90 95
Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser
100 105 110
Leu Ser Leu Ser Leu Gly Lys
115
<210> 72
<211> 229
<212> PRT
<213> Homo Sapiens (Homo Sapiens)
<220>
<223> hinge-CH 2-CH3
Spacer
<400> 72
Glu Ser Lys Tyr Gly Pro Pro Cys Pro Pro Cys Pro Ala Pro Glu Phe
1 5 10 15
Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr
20 25 30
Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val
35 40 45
Ser Gln Glu Asp Pro Glu Val Gln Phe Asn Trp Tyr Val Asp Gly Val
50 55 60
Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser
65 70 75 80
Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu
85 90 95
Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Gly Leu Pro Ser
100 105 110
Ser Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro
115 120 125
Gln Val Tyr Thr Leu Pro Pro Ser Gln Glu Glu Met Thr Lys Asn Gln
130 135 140
Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala
145 150 155 160
Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr
165 170 175
Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu
180 185 190
Thr Val Asp Lys Ser Arg Trp Gln Glu Gly Asn Val Phe Ser Cys Ser
195 200 205
Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser
210 215 220
Leu Ser Leu Gly Lys
225
<210> 73
<211> 200
<212> PRT
<213> Homo Sapiens (Homo Sapiens)
<220>
<223> IgD-hinge-Fc
<400> 73
Arg Trp Pro Glu Ser Pro Lys Ala Gln Ala Ser Ser Val Pro Thr Ala
1 5 10 15
Gln Pro Gln Ala Glu Gly Ser Leu Ala Lys Ala Thr Thr Ala Pro Ala
20 25 30
Thr Thr Arg Asn Thr Gly Arg Gly Gly Glu Glu Lys Lys Lys Glu Lys
35 40 45
Glu Lys Glu Glu Gln Glu Glu Arg Glu Thr Lys Thr Pro Glu Cys Pro
50 55 60
Ser His Thr Gln Pro Leu Gly Val Tyr Leu Leu Thr Pro Ala Val Gln
65 70 75 80
Asp Leu Trp Leu Arg Asp Lys Ala Thr Phe Thr Cys Phe Val Val Gly
85 90 95
Ser Asp Leu Lys Asp Ala His Leu Thr Trp Glu Val Ala Gly Lys Val
100 105 110
Pro Thr Gly Gly Val Glu Glu Gly Leu Leu Glu Arg His Ser Asn Gly
115 120 125
Ser Gln Ser Gln His Ser Arg Leu Thr Leu Pro Arg Ser Leu Trp Asn
130 135 140
Ala Gly Thr Ser Val Thr Cys Thr Leu Asn His Pro Ser Leu Pro Pro
145 150 155 160
Gln Arg Leu Met Ala Leu Arg Glu Pro Ala Ala Gln Ala Pro Val Lys
165 170 175
Leu Ser Leu Asn Leu Leu Ala Ser Ser Asp Pro Pro Glu Ala Ala Ser
180 185 190
Trp Leu Leu Cys Glu Val Ser Gly
195 200
<210> 74
<211> 14
<212> PRT
<213> artificial sequence
<220>
<223> exemplary IgG
Hinge
<400> 74
Glu Val Val Val Lys Tyr Gly Pro Pro Cys Pro Pro Cys Pro
1 5 10
<210> 75
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> exemplary IgG
Hinge
<220>
<221> variant
<222> 1
<223> Xaa is glycine, cysteine or arginine
<220>
<221> variant
<222> 4
<223> Xaa is cysteine or threonine
<400> 75
Xaa Pro Pro Xaa Pro
1 5
<210> 76
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> exemplary IgG
Hinge
<400> 76
Glu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro
1 5 10 15
<210> 77
<211> 12
<212> PRT
<213> artificial sequence
<220>
<223> exemplary IgG
Hinge
<400> 77
Glu Arg Lys Cys Cys Val Glu Cys Pro Pro Cys Pro
1 5 10
<210> 78
<211> 61
<212> PRT
<213> artificial sequence
<220>
<223> exemplary IgG
Hinge
<400> 78
Glu Leu Lys Thr Pro Leu Gly Asp Thr His Thr Cys Pro Arg Cys Pro
1 5 10 15
Glu Pro Lys Ser Cys Asp Thr Pro Pro Pro Cys Pro Arg Cys Pro Glu
20 25 30
Pro Lys Ser Cys Asp Thr Pro Pro Pro Cys Pro Arg Cys Pro Glu Pro
35 40 45
Lys Ser Cys Asp Thr Pro Pro Pro Cys Pro Arg Cys Pro
50 55 60
<210> 79
<211> 12
<212> PRT
<213> artificial sequence
<220>
<223> exemplary IgG
Hinge
<400> 79
Glu Ser Lys Tyr Gly Pro Pro Cys Pro Ser Cys Pro
1 5 10
<210> 80
<211> 12
<212> PRT
<213> artificial sequence
<220>
<223> exemplary IgG
Hinge
<400> 80
Glu Ser Lys Tyr Gly Pro Pro Cys Pro Pro Cys Pro
1 5 10
<210> 81
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> exemplary IgG
Hinge
<400> 81
Tyr Gly Pro Pro Cys Pro Pro Cys Pro
1 5
<210> 82
<211> 10
<212> PRT
<213> artificial sequence
<220>
<223> exemplary IgG
Hinge
<400> 82
Lys Tyr Gly Pro Pro Cys Pro Pro Cys Pro
1 5 10
<210> 83
<211> 27
<212> PRT
<213> Homo Sapiens (Homo Sapiens)
<220>
<223> CD28 (accession number)
P10747
Amino acids
153-179 )
<400> 83
Phe Trp Val Leu Val Val Val Gly Gly Val Leu Ala Cys Tyr Ser Leu
1 5 10 15
Leu Val Thr Val Ala Phe Ile Ile Phe Trp Val
20 25
<210> 84
<211> 66
<212> PRT
<213> Homo Sapiens (Homo Sapiens)
<220>
<223> CD28 (accession number)
P10747
Amino acids
114-179)
<400> 84
Ile Glu Val Met Tyr Pro Pro Pro Tyr Leu Asp Asn Glu Lys Ser Asn
1 5 10 15
Gly Thr Ile Ile His Val Lys Gly Lys His Leu Cys Pro Ser Pro Leu
20 25 30
Phe Pro Gly Pro Ser Lys Pro Phe Trp Val Leu Val Val Val Gly Gly
35 40 45
Val Leu Ala Cys Tyr Ser Leu Leu Val Thr Val Ala Phe Ile Ile Phe
50 55 60
Trp Val
65
<210> 85
<211> 41
<212> PRT
<213> Homo Sapiens (Homo Sapiens)
<220>
<223> CD28 (P10747)
Amino acids
180-220)
<400> 85
Arg Ser Lys Arg Ser Arg Leu Leu His Ser Asp Tyr Met Asn Met Thr
1 5 10 15
Pro Arg Arg Pro Gly Pro Thr Arg Lys His Tyr Gln Pro Tyr Ala Pro
20 25 30
Pro Arg Asp Phe Ala Ala Tyr Arg Ser
35 40
<210> 86
<211> 41
<212> PRT
<213> Homo Sapiens (Homo Sapiens)
<220>
<223> CD28 (LL to GG)
<400> 86
Arg Ser Lys Arg Ser Arg Gly Gly His Ser Asp Tyr Met Asn Met Thr
1 5 10 15
Pro Arg Arg Pro Gly Pro Thr Arg Lys His Tyr Gln Pro Tyr Ala Pro
20 25 30
Pro Arg Asp Phe Ala Ala Tyr Arg Ser
35 40
<210> 87
<211> 42
<212> PRT
<213> Homo Sapiens (Homo Sapiens)
<220>
<223> 4-1BB (Q07011.1)
Amino acids
214-255)
<400> 87
Lys Arg Gly Arg Lys Lys Leu Leu Tyr Ile Phe Lys Gln Pro Phe Met
1 5 10 15
Arg Pro Val Gln Thr Thr Gln Glu Glu Asp Gly Cys Ser Cys Arg Phe
20 25 30
Pro Glu Glu Glu Glu Gly Gly Cys Glu Leu
35 40
<210> 88
<211> 112
<212> PRT
<213> Homo Sapiens (Homo Sapiens)
<220>
<223> CD3ζ
<400> 88
Arg Val Lys Phe Ser Arg Ser Ala Asp Ala Pro Ala Tyr Gln Gln Gly
1 5 10 15
Gln Asn Gln Leu Tyr Asn Glu Leu Asn Leu Gly Arg Arg Glu Glu Tyr
20 25 30
Asp Val Leu Asp Lys Arg Arg Gly Arg Asp Pro Glu Met Gly Gly Lys
35 40 45
Pro Arg Arg Lys Asn Pro Gln Glu Gly Leu Tyr Asn Glu Leu Gln Lys
50 55 60
Asp Lys Met Ala Glu Ala Tyr Ser Glu Ile Gly Met Lys Gly Glu Arg
65 70 75 80
Arg Arg Gly Lys Gly His Asp Gly Leu Tyr Gln Gly Leu Ser Thr Ala
85 90 95
Thr Lys Asp Thr Tyr Asp Ala Leu His Met Gln Ala Leu Pro Pro Arg
100 105 110
<210> 89
<211> 112
<212> PRT
<213> Homo Sapiens (Homo Sapiens)
<220>
<223> CD3ζ
<400> 89
Arg Val Lys Phe Ser Arg Ser Ala Glu Pro Pro Ala Tyr Gln Gln Gly
1 5 10 15
Gln Asn Gln Leu Tyr Asn Glu Leu Asn Leu Gly Arg Arg Glu Glu Tyr
20 25 30
Asp Val Leu Asp Lys Arg Arg Gly Arg Asp Pro Glu Met Gly Gly Lys
35 40 45
Pro Arg Arg Lys Asn Pro Gln Glu Gly Leu Tyr Asn Glu Leu Gln Lys
50 55 60
Asp Lys Met Ala Glu Ala Tyr Ser Glu Ile Gly Met Lys Gly Glu Arg
65 70 75 80
Arg Arg Gly Lys Gly His Asp Gly Leu Tyr Gln Gly Leu Ser Thr Ala
85 90 95
Thr Lys Asp Thr Tyr Asp Ala Leu His Met Gln Ala Leu Pro Pro Arg
100 105 110
<210> 90
<211> 112
<212> PRT
<213> Homo Sapiens (Homo Sapiens)
<220>
<223> CD3ζ
<400> 90
Arg Val Lys Phe Ser Arg Ser Ala Asp Ala Pro Ala Tyr Lys Gln Gly
1 5 10 15
Gln Asn Gln Leu Tyr Asn Glu Leu Asn Leu Gly Arg Arg Glu Glu Tyr
20 25 30
Asp Val Leu Asp Lys Arg Arg Gly Arg Asp Pro Glu Met Gly Gly Lys
35 40 45
Pro Arg Arg Lys Asn Pro Gln Glu Gly Leu Tyr Asn Glu Leu Gln Lys
50 55 60
Asp Lys Met Ala Glu Ala Tyr Ser Glu Ile Gly Met Lys Gly Glu Arg
65 70 75 80
Arg Arg Gly Lys Gly His Asp Gly Leu Tyr Gln Gly Leu Ser Thr Ala
85 90 95
Thr Lys Asp Thr Tyr Asp Ala Leu His Met Gln Ala Leu Pro Pro Arg
100 105 110
<210> 91
<211> 30
<212> PRT
<213> artificial sequence
<220>
<223> linker; p is proline, G is glycine and S is
Serine (serine)
<400> 91
Pro Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly
1 5 10 15
Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Gly Gly Pro
20 25 30
<210> 92
<211> 17
<212> PRT
<213> artificial sequence
<220>
<223> joint
<400> 92
Gly Ser Ala Asp Asp Ala Lys Lys Asp Ala Ala Lys Lys Asp Gly Lys
1 5 10 15
Ser

Claims (65)

1. A method of determining the efficacy of a therapeutic cell composition, the method comprising:
performing a plurality of incubations, each of the plurality of incubations comprising culturing cells of a therapeutic cell composition comprising cells engineered to express a recombinant receptor with a recombinant receptor stimulator, wherein:
binding of the recombinant receptor stimulator to the recombinant receptor stimulates recombinant receptor-dependent activity of the cell; and is also provided with
Each of the plurality of incubations includes a different stepwise adjustment ratio of cells of the therapeutic cell composition to the recombinant receptor stimulator;
Measuring recombinant receptor-dependent activity from each of the plurality of incubations; and
based on the recombinant receptor-dependent activity measured from each of the plurality of incubations, a stepwise adjustment ratio is determined that results in a half-maximal recombinant receptor-dependent activity.
2. The method of claim 1, further comprising determining the relative efficacy of the therapeutic cell composition by comparing the stepwise adjustment ratio of the half-maximal recombinant receptor-dependent activity resulting in the therapeutic cell composition to the stepwise adjustment ratio of the half-maximal recombinant receptor-dependent activity resulting in a reference standard.
3. A method of determining the efficacy of a therapeutic cell composition, the method comprising:
performing a plurality of incubations, each of the plurality of incubations comprising culturing cells of a therapeutic cell composition comprising cells engineered to express a recombinant receptor with a recombinant receptor stimulator, wherein:
binding of the recombinant receptor stimulator to the recombinant receptor stimulates recombinant receptor-dependent activity of the cell; and is also provided with
Each of the plurality of incubations includes a different stepwise adjustment ratio of cells of the therapeutic cell composition to the recombinant receptor stimulator;
Measuring recombinant receptor-dependent activity from each of the plurality of incubations; and
the relative potency of the therapeutic cell composition is determined by comparing the half maximal recombinant receptor-dependent activity of the therapeutic cell composition to the half maximal recombinant receptor-dependent activity of a reference standard.
4. The method of any one of claims 1-3, wherein each of the plurality of incubations comprises culturing a constant number of cells of the therapeutic composition with different amounts of the recombinant receptor stimulator to generate a plurality of different stepwise adjustment ratios.
5. The method of any one of claims 1-3, wherein each of the plurality of incubations comprises culturing a constant amount of recombinant receptor stimulator with a different number of cells of the therapeutic composition to generate a plurality of different stepwise adjustment ratios.
6. The method of any one of claims 1-5, wherein the plurality of incubations is performed on two or more, optionally 3, 4, 5, 6, 7, 8, 9, 10 or more therapeutic cell compositions.
7. The method of claim 6, wherein the two or more therapeutic cell compositions each comprise the same recombinant receptor.
8. The method of claim 6, wherein the two or more therapeutic cell compositions each comprise a different recombinant receptor.
9. The method of claim 6, wherein at least one of the two or more therapeutic cell compositions comprises a different recombinant receptor than the other therapeutic composition.
10. The method of any one of claims 6-9, wherein each of the two or more therapeutic cell compositions are manufactured using the same manufacturing process.
11. The method of any one of claims 6-9, wherein the two or more therapeutic cell compositions are each manufactured using a different manufacturing process.
12. The method of any one of claims 6-9, wherein at least one of the two or more therapeutic cell compositions is manufactured using a manufacturing process that is different from a manufacturing process used to manufacture other therapeutic cell compositions.
13. The method of any one of claims 6-12, wherein the two or more therapeutic cell compositions are produced by cells from a single subject.
14. The method of any one of claims 6-12, wherein the two or more therapeutic cell compositions are produced by cells from different subjects.
15. The method of claim 13, wherein the subject is a healthy subject or a subject with a disease or disorder.
16. The method of claim 14, wherein each of the different subjects has the same disease or disorder.
17. The method of claim 14, wherein each of the different subjects is to be treated with the same therapeutic cell composition to treat the disease or disorder in the subject.
18. The method of any one of claims 1-17, wherein the plurality of incubations is at least three incubations.
19. The method of any one of claims 1-18, wherein the plurality of incubations is at least five incubations.
20. The method of any one of claims 1-19, wherein the plurality of incubations is at least seven incubations.
21. The method of any one of claims 1-20, wherein the plurality of incubations is at least ten incubations.
22. The method of any one of claims 1-21, wherein the recombinant receptor-dependent activity comprises one or more of: cytokine expression, cytolytic activity, receptor up-regulation, receptor down-regulation, proliferation, gene up-regulation, gene down-regulation, or cellular health.
23. The method of any one of claims 1-22, wherein the recombinant receptor-dependent activity comprises or is cytokine expression or production.
24. The method of any one of claims 1-23, wherein the recombinant receptor-dependent activity comprises or is cytokine expression or production, wherein the cytokine is TNF-a, IFN- γ (IFNg), or IL-2.
25. The method of any one of claims 1-24, wherein the recombinant receptor-dependent activity comprises or is a cytolytic activity.
26. The method of any one of claims 1-25, wherein the recombinant receptor-dependent activity comprises or is receptor up-regulation.
27. The method of any one of claims 1-26, wherein the recombinant receptor-dependent activity comprises or is receptor down-regulation.
28. The method of any one of claims 1-27, wherein the recombinant receptor-dependent activity comprises or is proliferation.
29. The method of any one of claims 1-28, wherein the recombinant receptor-dependent activity comprises or is gene up-regulation.
30. The method of any one of claims 1-29, wherein the recombinant receptor-dependent activity comprises or is down-regulated.
31. The method of any one of claims 1-30, wherein the recombinant receptor-dependent activity comprises or is cellular health.
32. The method of any one of claims 1-31, wherein the recombinant receptor-dependent activity comprises or is a cellular health, wherein the cellular health comprises one or more of cell death, cell diameter, viable cell concentration, and cell count.
33. The method of any one of claims 1-32, wherein the recombinant receptor-dependent activity measured at each of the plurality of incubations is normalized to a maximum receptor-dependent activity measured for the therapeutic cell composition.
34. The method of any one of claims 1-33, wherein the reference standard is a therapeutic cell composition comprising a validated step-by-step adjustment ratio that results in half-maximal recombinant receptor-dependent activity, a commercially available therapeutic cell composition, a therapeutic cell composition manufactured using the same manufacturing process as that used to manufacture the therapeutic cell composition, a therapeutic cell composition manufactured using a different manufacturing process than that used to manufacture the therapeutic cell composition, a therapeutic cell composition comprising the same recombinant receptor as that therapeutic cell composition, a therapeutic cell composition comprising a different recombinant receptor than that therapeutic cell composition, a therapeutic cell composition manufactured from the same subject, or a therapeutic cell composition manufactured from a different subject.
35. The method of any one of claims 6-34, wherein the reference standard is one of the two or more therapeutic compositions.
36. The method of any one of claims 1-35, wherein the recombinant receptor stimulator comprises a target antigen of the recombinant receptor or an extracellular domain-binding portion thereof, optionally a recombinant antigen.
37. The method of claim 36, wherein the recombinant receptor stimulant comprises an ectodomain binding portion of the antigen, and the ectodomain binding portion comprises an epitope recognized by the recombinant receptor.
38. The method of any one of claims 1-35, wherein the recombinant receptor stimulant is an antibody specific for an extracellular binding domain of the recombinant receptor.
39. The method of any one of claims 1-35 and 38, wherein the recombinant receptor stimulant is an anti-idiotype antibody specific for an extracellular antigen-binding domain of the recombinant receptor.
40. The method of any one of claims 1-39, wherein the recombinant receptor stimulant is immobilized or attached to a solid support.
41. The method of claim 40, wherein the solid support is a surface of a vessel, optionally a well of a microplate, in which the plurality of incubations are performed.
42. The method of claim 40, wherein the solid support is a bead.
43. The method of any one of claims 1-35, wherein the recombinant receptor stimulant is an antigen-expressing cell, optionally wherein the cell is a clone, derived from a cell line, or a primary cell taken from a subject.
44. The method of claim 43, wherein the antigen expressing cell is a cell line.
45. The method of claim 44, wherein the cell line is a tumor cell line.
46. The method of claim 43, wherein the antigen expressing cell is a cell that has been introduced, optionally by transduction, to express an antigen of the recombinant receptor.
47. The method of any one of claims 1-46, wherein the stepwise adjustment ratio reaches a linear dose response range of recombinant receptor-dependent activity of the reference standard.
48. The method of claim 47, wherein the stepwise adjustment ratio comprises a lower asymptote (minimum) and a higher asymptote (maximum) recombinant receptor-dependent activity of the reference standard.
49. The method of any one of claims 1-37, wherein the therapeutic cell composition comprises a single cell subtype enriched or purified from a biological sample or a mixed population of cell subtypes obtained optionally by mixing cell subtypes enriched or purified from a biological sample.
50. The method of claim 49, wherein the biological sample comprises a whole blood sample, a buffy coat sample, a Peripheral Blood Mononuclear Cell (PBMC) sample, an unfractionated cell sample, a lymphocyte sample, a leukocyte sample, a apheresis product, or a leukocyte apheresis product.
51. The method of any one of claims 1-50, wherein the therapeutic cell composition comprises primary cells.
52. The method of any one of claims 1-51, wherein the therapeutic cell composition comprises autologous cells from the subject to be treated.
53. The method of any one of claims 1-52, wherein the therapeutic cell composition comprises allogeneic cells.
54. The method of any one of claims 1-53, wherein the therapeutic cell composition comprises cd3+, cd4+, and/or cd8+ T cells.
55. The method of any one of claims 1-54, wherein the therapeutic cell composition comprises cd4+ T cells and cd8+ T cells.
56. The method of any one of claims 1-55, wherein the recombinant receptor is a Chimeric Antigen Receptor (CAR).
57. The method of any one of claims 1-56, wherein the plurality of incubations are performed in a flask, tube, or multi-well plate.
58. The method of any one of claims 1-57, wherein each of the plurality of incubations is performed separately in a well of a multi-well plate.
59. The method of claim 57 or claim 58, wherein the multi-well plate is a 96-well plate, a 48-well plate, a 12-well plate, or a 6-well plate.
60. The method of any one of claims 1 and 4-59, further comprising determining a dose of cells of the therapeutic composition for administration to a subject in need thereof based on a stepwise adjustment ratio that results in half-maximal recombinant receptor-dependent activity.
61. The method of any one of claims 2-59, further comprising determining a dose of cells of the therapeutic composition for administration to a subject in need thereof based on the relative potency.
62. The method of claim 60 or claim 61, wherein the subject has a disease or disorder.
63. The method of any one of claims 15-62, wherein the disease or disorder is cancer.
64. The method of any one of claims 2-63, further comprising determining a manufacturing process that produces an optimal therapeutic cellular composition efficacy based on the relative efficacy, wherein the optimal therapeutic cellular composition efficacy is associated with a complete and/or sustained response and/or reduced toxicity.
65. The method of any one of claims 2-64, further comprising determining a manufacturing process that produces a therapeutic cell composition having a reduced or low difference in potency based on the relative potency, wherein the reduced or low difference is determined by comparison with a difference in a different manufacturing process.
CN202280036193.9A 2021-03-22 2022-03-21 Method for determining the efficacy of therapeutic cell compositions Pending CN117321417A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US202163164527P 2021-03-22 2021-03-22
US63/164,527 2021-03-22
PCT/US2022/021225 WO2022204070A1 (en) 2021-03-22 2022-03-21 Methods of determining potency of a therapeutic cell composition

Publications (1)

Publication Number Publication Date
CN117321417A true CN117321417A (en) 2023-12-29

Family

ID=81327916

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280036193.9A Pending CN117321417A (en) 2021-03-22 2022-03-21 Method for determining the efficacy of therapeutic cell compositions

Country Status (8)

Country Link
US (1) US20240168012A1 (en)
EP (1) EP4314814A1 (en)
JP (1) JP2024511418A (en)
KR (1) KR20230159851A (en)
CN (1) CN117321417A (en)
AU (1) AU2022241654A1 (en)
CA (1) CA3210581A1 (en)
WO (1) WO2022204070A1 (en)

Family Cites Families (108)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5876435A (en) 1981-10-30 1983-05-09 Japan Synthetic Rubber Co Ltd Polymeric particle
US4452773A (en) 1982-04-05 1984-06-05 Canadian Patents And Development Limited Magnetic iron-dextran microspheres
NO155316C (en) 1982-04-23 1987-03-11 Sintef PROCEDURE FOR MAKING MAGNETIC POLYMER PARTICLES.
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4554088A (en) 1983-05-12 1985-11-19 Advanced Magnetics Inc. Magnetic particles for use in separations
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
US4690915A (en) 1985-08-08 1987-09-01 The United States Of America As Represented By The Department Of Health And Human Services Adoptive immunotherapy as a treatment modality in humans
US4795698A (en) 1985-10-04 1989-01-03 Immunicon Corporation Magnetic-polymer particles
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
US6548640B1 (en) 1986-03-27 2003-04-15 Btg International Limited Altered antibodies
IN165717B (en) 1986-08-07 1989-12-23 Battelle Memorial Institute
US5219740A (en) 1987-02-13 1993-06-15 Fred Hutchinson Cancer Research Center Retroviral gene transfer into diploid fibroblasts for gene therapy
JP3101690B2 (en) 1987-03-18 2000-10-23 エス・ビィ・2・インコーポレイテッド Modifications of or for denatured antibodies
US5091206A (en) 1987-10-26 1992-02-25 Baxter Diagnostics Inc. Process for producing magnetically responsive polymer particles and application thereof
US5395688A (en) 1987-10-26 1995-03-07 Baxter Diagnostics Inc. Magnetically responsive fluorescent polymer particles
ATE114507T1 (en) 1988-12-28 1994-12-15 Stefan Miltenyi METHODS AND MATERIALS FOR HIGH GRADUATION MAGNETIC SEPARATION OF BIOLOGICAL MATERIALS.
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
FR2645160B1 (en) 1989-03-31 1992-10-02 Rhone Poulenc Chimie
FR2656318B1 (en) 1989-12-27 1994-02-04 Rhone Poulenc Chimie MAGNETISABLE CORE-SHELL MICROSPHERES BASED ON CROSSLINKED ORGANOPOLYSILOXANE, THEIR PREPARATION PROCESS AND THEIR APPLICATION IN BIOLOGY.
US5318797A (en) 1990-06-20 1994-06-07 Clarkson University Coated particles, hollow particles, and process for manufacturing the same
US5200084A (en) 1990-09-26 1993-04-06 Immunicon Corporation Apparatus and methods for magnetic separation
DE69128037T2 (en) 1990-11-13 1998-05-07 Immunex Corp BIFUNCTIONAL SELECTABLE FUSION GENES
EP1400536A1 (en) 1991-06-14 2004-03-24 Genentech Inc. Method for making humanized antibodies
DE4228458A1 (en) 1992-08-27 1994-06-01 Beiersdorf Ag Multicistronic expression units and their use
AU6953394A (en) 1993-05-21 1994-12-20 Targeted Genetics Corporation Bifunctional selectable fusion genes based on the cytosine deaminase (cd) gene
US5827642A (en) 1994-08-31 1998-10-27 Fred Hutchinson Cancer Research Center Rapid expansion method ("REM") for in vitro propagation of T lymphocytes
WO1996013593A2 (en) 1994-10-26 1996-05-09 Procept, Inc. Soluble single chain t cell receptors
WO1996018105A1 (en) 1994-12-06 1996-06-13 The President And Fellows Of Harvard College Single chain t-cell receptor
US20020150914A1 (en) 1995-06-30 2002-10-17 Kobenhavns Universitet Recombinant antibodies from a phage display library, directed against a peptide-MHC complex
US6013516A (en) 1995-10-06 2000-01-11 The Salk Institute For Biological Studies Vector and method of use for nucleic acid delivery to non-dividing cells
US5834121A (en) 1996-01-16 1998-11-10 Solid Phase Sciences Corp. Composite magnetic beads
DE19608753C1 (en) 1996-03-06 1997-06-26 Medigene Gmbh Transduction system based on rep-negative adeno-associated virus vector
US6451995B1 (en) 1996-03-20 2002-09-17 Sloan-Kettering Institute For Cancer Research Single chain FV polynucleotide or peptide constructs of anti-ganglioside GD2 antibodies, cells expressing same and related methods
DE69605062T2 (en) 1996-04-24 2000-07-13 Claude Fell CELL SEPARATION DEVICE FOR BIOLOGICAL LIQUIDS LIKE BLOOD
WO1998040510A1 (en) 1997-03-11 1998-09-17 Regents Of The University Of Minnesota Dna-based transposon system for the introduction of nucleic acid into dna of a cell
ATE533784T1 (en) 1997-10-02 2011-12-15 Altor Bioscience Corp SOLUBLE, SINGLE-CHAIN T-CELL RECEPTOR PROTEINS
US6074884A (en) 1997-10-09 2000-06-13 Coulter International Corp. Stable protein-nickel particles and methods of production and use thereof
CA2309000A1 (en) 1997-11-13 1999-05-27 Regents Of The University Of Minnesota Tc1-based transposon vectors
EP1034298B1 (en) 1997-12-05 2011-11-02 The Scripps Research Institute Humanization of murine antibody
US5994136A (en) 1997-12-12 1999-11-30 Cell Genesys, Inc. Method and means for producing high titer, safe, recombinant lentivirus vectors
GB9809951D0 (en) 1998-05-08 1998-07-08 Univ Cambridge Tech Binding molecules
EA200001216A1 (en) 1998-05-19 2001-06-25 Авидекс Лимитед SOLUBLE T-LYMPHOCYTIC RECEPTOR
WO2000014257A1 (en) 1998-09-04 2000-03-16 Sloan-Kettering Institute For Cancer Research Fusion receptors specific for prostate-specific membrane antigen and uses thereof
US6410319B1 (en) 1998-10-20 2002-06-25 City Of Hope CD20-specific redirected T cells and their use in cellular immunotherapy of CD20+ malignancies
CN1182882C (en) 1998-12-24 2005-01-05 生物安全股份有限公司 Blood separation system, in particular for concentrating hematopoietic stem cells
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US20040191260A1 (en) 2003-03-26 2004-09-30 Technion Research & Development Foundation Ltd. Compositions capable of specifically binding particular human antigen presenting molecule/pathogen-derived antigen complexes and uses thereof
AU2001265346A1 (en) 2000-06-02 2001-12-17 Memorial Sloan-Kettering Cancer Center Artificial antigen presenting cells and methods of use thereof
EP1334188B1 (en) 2000-11-07 2006-08-30 City of Hope Cd19-specific redirected immune cells
US7070995B2 (en) 2001-04-11 2006-07-04 City Of Hope CE7-specific redirected immune cells
US20090257994A1 (en) 2001-04-30 2009-10-15 City Of Hope Chimeric immunoreceptor useful in treating human cancers
PL208712B1 (en) 2001-08-31 2011-05-31 Avidex Ltd Soluble t cell receptor
US7939059B2 (en) 2001-12-10 2011-05-10 California Institute Of Technology Method for the generation of antigen-specific lymphocytes
US6992176B2 (en) 2002-02-13 2006-01-31 Technion Research & Development Foundation Ltd. Antibody having a T-cell receptor-like specificity, yet higher affinity, and the use of same in the detection and treatment of cancer, viral infection and autoimmune disease
AU2003216341A1 (en) 2002-02-20 2003-09-09 Dyax Corporation Mhc-peptide complex binding ligands
US20030170238A1 (en) 2002-03-07 2003-09-11 Gruenberg Micheal L. Re-activated T-cells for adoptive immunotherapy
US7446190B2 (en) 2002-05-28 2008-11-04 Sloan-Kettering Institute For Cancer Research Nucleic acids encoding chimeric T cell receptors
NZ539225A (en) 2002-10-09 2006-09-29 Avidex Ltd Single chain recombinant T cell receptors
US20050129671A1 (en) 2003-03-11 2005-06-16 City Of Hope Mammalian antigen-presenting T cells and bi-specific T cells
DK1631788T3 (en) 2003-05-16 2007-07-23 Univ Bruxelles Digital holographic microscope for 3D image generation and a process for its use
US20090226474A1 (en) 2004-05-27 2009-09-10 Weidanz Jon A Antibodies as T cell receptor mimics, methods of production and uses thereof
US20090304679A1 (en) 2004-05-27 2009-12-10 Weidanz Jon A Antibodies as T cell receptor mimics, methods of production and uses thereof
EP1773383B1 (en) 2004-05-27 2012-09-12 Jon A. Weidanz Antibodies as t cell receptor mimics, methods of production and uses thereof
DE602005022595D1 (en) 2004-06-29 2010-09-09 Immunocore Ltd A MODIFIED T-CELL RECEPTOR EXPRESSING CELLS
WO2006100651A1 (en) 2005-03-23 2006-09-28 Biosafe S.A. Integrated system for collecting, processing and transplanting cell subsets, including adult stem cells, for regenerative medicine
JP2011503244A (en) 2006-12-21 2011-01-27 インヴィトロジェン ダイナル エーエス Particles and their use in nucleic acid isolation methods or phosphorylated protein isolation methods
CA2967847C (en) 2007-03-30 2023-08-01 Memorial Sloan-Kettering Cancer Center Constitutive expression of costimulatory ligands on adoptively transferred t lymphocytes
JP5456689B2 (en) 2007-12-07 2014-04-02 ミルテンイ バイオテック ゲーエムベーハー A centrifuge that separates a sample into at least two components
US8479118B2 (en) 2007-12-10 2013-07-02 Microsoft Corporation Switching search providers within a browser search box
WO2009076524A2 (en) 2007-12-11 2009-06-18 The University Of North Carolina At Chapel Hill Polypurine tract modified retroviral vectors
JP5173594B2 (en) 2008-05-27 2013-04-03 キヤノン株式会社 Management apparatus, image forming apparatus, and processing method thereof
CA2735456C (en) 2008-08-26 2021-11-16 City Of Hope Method and compositions for enhanced anti-tumor effector functioning of t cells
CN102421801B (en) 2009-03-10 2016-03-16 比奥根Ma公司 Anti-BCMA antibody
US10464987B2 (en) 2009-10-06 2019-11-05 Abbvie Inc. Human single-chain T cell receptors
AU2010315243B2 (en) 2009-11-03 2016-08-25 City Of Hope Truncated epidermal growth factor receptor (EGFRt) for transduced T cell selection
MX347078B (en) 2010-12-09 2017-04-10 Univ Pennsylvania Use of chimeric antigen receptor-modified t cells to treat cancer.
AU2011343887B2 (en) 2010-12-14 2016-06-16 University Of Maryland, Baltimore Universal anti-tag chimeric antigen receptor-expressing T cells and methods of treating cancer
NZ743310A (en) 2011-03-23 2022-11-25 Fred Hutchinson Cancer Center Method and compositions for cellular immunotherapy
US9074000B2 (en) 2011-04-01 2015-07-07 Memorial Sloan Kettering Cancer Center T cell receptor-like antibodies specific for a WT1 peptide presented by HLA-A2
US8398282B2 (en) 2011-05-12 2013-03-19 Delphi Technologies, Inc. Vehicle front lighting assembly and systems having a variable tint electrowetting element
CN103827889B (en) 2011-07-19 2018-01-02 奥维茨奥成像系统公司 Method and system for the cancer cell in the cell sample that detects and/or classify
WO2013044225A1 (en) 2011-09-22 2013-03-28 The Trustees Of The University Of Pennsylvania A universal immune receptor expressed by t cells for the targeting of diverse and multiple antigens
AU2012335073B2 (en) 2011-11-11 2017-08-17 Fred Hutchinson Cancer Center Cyclin A1-targeted T-cell immunotherapy for cancer
AU2013221672B2 (en) 2012-02-13 2017-11-09 Seattle Children's Hospital D/B/A Seattle Children's Research Institute Bispecific chimeric antigen receptors and therapeutic uses thereof
WO2013126726A1 (en) 2012-02-22 2013-08-29 The Trustees Of The University Of Pennsylvania Double transgenic t cells comprising a car and a tcr and their methods of use
ES2858248T3 (en) 2012-05-03 2021-09-29 Hutchinson Fred Cancer Res Affinity Enhanced T Cell Receptors and Methods for Making Them
WO2013188864A2 (en) 2012-06-15 2013-12-19 Sinomab Bioscience Limited Anti-cd22 anti-idiotypic antibodies and uses thereof
JP6574381B2 (en) 2012-08-20 2019-09-11 フレッド ハッチンソン キャンサー リサーチ センター Methods and compositions for cellular immunotherapy
EP2898310B1 (en) 2012-09-20 2019-05-01 Ovizio Imaging Systems NV/SA Digital holographic microscope with fluid systems
CN104853766A (en) 2012-10-02 2015-08-19 纪念斯隆-凯特琳癌症中心 Compositions and methods for immunotherapy
UY35468A (en) 2013-03-16 2014-10-31 Novartis Ag CANCER TREATMENT USING AN ANTI-CD19 CHEMERIC ANTIGEN RECEIVER
CN111961647A (en) 2014-04-16 2020-11-20 朱诺治疗有限公司 Methods, kits and devices for expanding cell populations
ES2759260T3 (en) 2014-04-23 2020-05-08 Juno Therapeutics Inc Methods for Isolating, Cultivating, and Genetically Modifying Immune Cell Populations for Adoptive Therapy
CA2984484C (en) 2014-05-02 2024-01-23 The Trustees Of The University Of Pennsylvania Compositions and methods of chimeric autoantibody receptor t cells
TWI805109B (en) 2014-08-28 2023-06-11 美商奇諾治療有限公司 Antibodies and chimeric antigen receptors specific for cd19
ES2688035T3 (en) 2014-08-29 2018-10-30 Gemoab Monoclonals Gmbh Universal antigen receptor that expresses immune cells for addressing multiple multiple antigens, procedure for manufacturing it and using it for the treatment of cancer, infections and autoimmune diseases
CA2966538A1 (en) 2014-11-05 2016-05-12 Juno Therapeutics, Inc. Methods for transduction and cell processing
MX2017007242A (en) 2014-12-05 2018-01-25 Memorial Sloan Kettering Cancer Center Chimeric antigen receptors targeting g-protein coupled receptor and uses thereof.
FI3227432T3 (en) 2014-12-05 2024-01-09 Memorial Sloan Kettering Cancer Center Chimeric antigen receptors targeting b-cell maturation antigen and uses thereof
CN113429485A (en) 2014-12-05 2021-09-24 纪念斯隆-凯特琳癌症中心 Antibodies targeting B-cell maturation antigens and uses thereof
AU2015357535B2 (en) 2014-12-05 2020-05-14 Eureka Therapeutics, Inc. Antibodies targeting G-protein coupled receptor and methods of use
US11377481B2 (en) 2015-12-22 2022-07-05 The Trustees Of The University Of Pennsylvania SpyCatcher and SpyTag: universal immune receptors for T cells
BR112018070073A2 (en) 2016-04-01 2019-02-12 Kite Pharma, Inc. chimeric antigen and t-cell receptors and methods of use
KR20230107408A (en) 2016-07-29 2023-07-14 주노 쎄러퓨티크스 인코퍼레이티드 Anti-idiotypic antibodies against anti-cd19 antibodies
BR112019017767A2 (en) * 2017-02-27 2020-04-07 Juno Therapeutics Inc compositions, articles of manufacture and methods related to dosing in cell therapy
MX2020000900A (en) 2017-07-29 2021-01-08 Juno Therapeutics Inc Reagents for expanding cells expressing recombinant receptors.
MX2020004240A (en) 2017-11-01 2020-09-25 Juno Therapeutics Inc Process for generating therapeutic compositions of engineered cells.
US11859491B2 (en) 2018-04-09 2024-01-02 King Abdullah University Of Science And Technology Low power water cut sensing

Also Published As

Publication number Publication date
JP2024511418A (en) 2024-03-13
WO2022204070A1 (en) 2022-09-29
KR20230159851A (en) 2023-11-22
AU2022241654A1 (en) 2023-09-28
CA3210581A1 (en) 2022-09-29
AU2022241654A9 (en) 2024-02-22
US20240168012A1 (en) 2024-05-23
EP4314814A1 (en) 2024-02-07

Similar Documents

Publication Publication Date Title
US20190375815A1 (en) Treatment of cancer using chimeric t cell receptor proteins having multiple specificities
CN114761037A (en) Chimeric antigen receptor binding to BCMA and CD19 and uses thereof
CN117903316A (en) Anti-idiotype antibodies and related methods
JP2018519842A (en) Methods for improving the effectiveness and expansion of immune cells
US20200354677A1 (en) Process for generating therapeutic compositions of engineered cells
BR112020001719A2 (en) reagents for cell expansion that express recombinant receptors
EP3664820B1 (en) Methods for producing genetically engineered cell compositions and related compositions
JP2021532742A (en) T cells containing NEF and their production methods
CN115803824A (en) Methods of identifying characteristics associated with clinical response and uses thereof
US20210230671A1 (en) Methods for assessing integrated nucleic acids
WO2021183675A2 (en) Methods for generating engineered memory-like nk cells and compositions thereof
EP3704229B1 (en) Process for producing a t cell composition
CN112041432A (en) FOXP3 targeting agent compositions and methods of use for adoptive cell therapy
CN117321417A (en) Method for determining the efficacy of therapeutic cell compositions
TWI840351B (en) T cell receptors and engineered cells expressing same
RU2790444C2 (en) Methods for production of compositions of genetically engineered cells and related compositions
WO2024118469A1 (en) Humanized anti-hla-g chimeric antigen receptors and uses thereof
CN115427550A (en) T cell transduction methods
EA043641B1 (en) METHOD FOR OBTAINING T-CELL COMPOSITION

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination